US20100278822A1 - Stable high protein concentration formulations of human anti-tnf-alpha-antibodies - Google Patents

Stable high protein concentration formulations of human anti-tnf-alpha-antibodies Download PDF

Info

Publication number
US20100278822A1
US20100278822A1 US12/772,595 US77259510A US2010278822A1 US 20100278822 A1 US20100278822 A1 US 20100278822A1 US 77259510 A US77259510 A US 77259510A US 2010278822 A1 US2010278822 A1 US 2010278822A1
Authority
US
United States
Prior art keywords
formulation
antibody
seq
amino acid
acid sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/772,595
Other languages
English (en)
Inventor
Wolfgang Fraunhofer
Hans-Juergen Krause
Michael Neu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Biotechnology Ltd
Abbott GmbH and Co KG
Original Assignee
Abbott Biotech Ltd Bermuda
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbott Biotech Ltd Bermuda filed Critical Abbott Biotech Ltd Bermuda
Priority to US12/772,595 priority Critical patent/US20100278822A1/en
Assigned to ABBOTT GMBH & CO. KG reassignment ABBOTT GMBH & CO. KG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KRAUSE, HANS-JUERGEN, NEU, MICHAEL
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FRAUNHOFER, WOLFGANG
Assigned to ABBOTT GMBH & CO., KG reassignment ABBOTT GMBH & CO., KG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FRAUNHOFER, WOLFGANG
Assigned to ABBOTT BIOTECHNOLOGY LTD. reassignment ABBOTT BIOTECHNOLOGY LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT LABORATORIES
Assigned to ABBOTT BIOTECHNOLOGY LTD. reassignment ABBOTT BIOTECHNOLOGY LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT GMBH & CO. KG
Publication of US20100278822A1 publication Critical patent/US20100278822A1/en
Assigned to ABBVIE BIOTECHNOLOGY LTD. reassignment ABBVIE BIOTECHNOLOGY LTD. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT BIOTECHNOLOGY LTD.
Priority to US14/170,061 priority patent/US20140141008A1/en
Priority to US14/170,026 priority patent/US20140141007A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins

Definitions

  • therapeutic proteins such as antibodies
  • stability e.g., suitability for administration, concentration.
  • therapeutic proteins During manufacturing, storage, and delivery, therapeutic proteins have been known to undergo physical and chemical degradations. These instabilities can reduce the potency of the protein and increase the risk of adverse events in patients, and, therefore, significantly impact regulatory approval (see, e.g., Wang, et al. (2007) J Pharm Sci 96:1). As such, a stable protein formulation is essential to the success of a therapeutic protein.
  • High protein concentration formulations are desirable as they can impact the mode (e.g., intravenous vs. subcutaneous) and frequency of administration of the drug to a subject.
  • the present invention is based, at least in part, on the discovery of new high-concentration formulations of human anti-TNF-a antibodies, or antigen-binding fragments thereof, e.g., adalimumab.
  • the formulations of the invention provide a number of surprising characteristics given the high concentration of antibody. For example, the formulations of the invention maintain physical and chemical stability over extended periods despite the high concentration of protein, and have a viscosity suitable for subcutaneous administration.
  • the formulations of the invention are established, at least in part, on the surprising finding that a human anti-TNF-alpha antibody, or antigen-binding portion thereof, can remain soluble at a high concentration (e.g., 100 mg/mL) and remain non-aggregated while maintaining a viscosity suitable for injection (e.g., subcutaneous administration).
  • the formulation of the present invention is also surprising in that a high concentration (e.g., 100 mg/mL) of human anti-TNF-alpha antibody, or antigen-binding portion thereof, can remain soluble and remain non-aggregated and chemically stable (e.g., no oxidation or deamidation) over a wide pH range, e.g., about pH 5.2 to about pH 6.0.
  • One aspect of the invention provides a liquid pharmaceutical formulation comprising more than 40 mg of a polyol and at least about 100 mg/mL of a human anti-TNF-alpha antibody, or antigen-binding portion thereof.
  • Another aspect of the invention provides a liquid pharmaceutical formulation comprising more than 20 mg of a polyol and at least about 100 mg/mL of a human anti-TNF-alpha antibody, or antigen-binding portion thereof.
  • the formulations of the invention do not contain NaCl.
  • the invention also features a liquid pharmaceutical formulation having a pH of about 5.0 to 6.4 and comprising at least about 100 mg/mL of a human anti-TNF-alpha antibody, or antigen-binding portion thereof, wherein the formulation does not contain NaCl and has a turbidity of less than 60 NTU after a standard 24 hour stir-stress assay or after 24 months of long-term storage as liquid.
  • the invention further provides a liquid pharmaceutical formulation having a pH of about 5.0 to 6.4 and comprising at least about 100 mg/mL of a human anti-TNF-alpha antibody, or antigen-binding portion thereof, wherein the formulation does not contain NaCl and has a turbidity of less than 100 NTU after a standard 48 hour stir-stress assay.
  • Another aspect of the invention includes a liquid pharmaceutical formulation having a pH of about 5.0 to 6.4 and comprising at least about 100 mg/mL of a human anti-TNF-alpha antibody, or antigen-binding portion thereof, wherein the formulation does not contain NaCl and has a turbidity of less than 40 NTU after 3 months storage at 5° C., 25° C., or 40° C.
  • the invention also provides a liquid pharmaceutical formulation comprising at least about 100 mg/mL of a human anti-TNF-alpha antibody, or antigen-binding portion thereof; more than about 20 mg/mL of a polyol; 0.1-2.0 mg/mL of a surfactant; about 1.15-1.45 mg/mL of citric acid * H 2 O; about 0.2-0.4 mg/mL of sodium citrate dehydrate; about 1.35-1.75 mg/mL of Na 2 HPO 4 * 2 H 2 O; about 0.75-0.95 mg/mL of NaH 2 PO 4 * 2H 2 O, wherein the formulation has a pH of about 4.7 to 6.5 and does not comprise NaCl.
  • the formulation of the invention is suitable for subcutaneous administration.
  • the invention also includes the use of the formulation of the invention comprising a human TNF alpha antibody, or antigen-binding portion thereof, for the treatment of a disorder associated with detrimental TNF alpha activity in a subject.
  • the formulation of the invention has a concentration of a human TNF alpha antibody, or antigen binding portion thereof, and a viscosity of between about 3.1-3.3 mPas*s.
  • the formulation of the invention comprises more than 20 mg of a polyol. Additional amounts of polyol which may be included in the formulation of the invention are more than 30 mg of the polyol. Alternatively, more than 40 mg of the polyol may be used in the formulation of the invention, including, but not limited to, 40-45 mg, or about 42 mg.
  • the polyol used in the formulation of the invention is a sugar alcohol, such as, but not limited to, mannitol or sorbitol.
  • the formulation comprises about 40-45 mg/mL of either mannitol or sorbitol.
  • the surfactant is polysorbate 80. In a further embodiment, about 0.1-2.0 mg/mL of polysorbate 80 is used in the formulation of the invention.
  • the formulation comprises about 1.30-1.31 mg/mL of citric acid * H 2 O.
  • the formulation comprises about 0.30-0.31 mg/mL sodium citrate dehydrate.
  • the formulation comprises about 1.50-1.56 mg/mL of Na 2 HPO 4 * 2H 2 O.
  • the formulation comprises about 0.83-0.89 mg/mL of NaH 2 PO 4 * 2H 2 O.
  • the pH of the formulation of the invention ranges from about 4.8 to about 6.4.
  • the pH of the formulation of the invention may range from either about 5.0 to about 5.4 (e.g., about 5.2) or may range from about 5.8 to about 6.4 (e.g., about 6.0).
  • An advantage of the formulation of the invention is that it provides a high concentration of antibody without increased protein aggregation, which commonly occurs with increased protein concentration.
  • the formulation of the invention has less than about 1% aggregate protein.
  • formulations described herein having a concentration of at least about 50 mg/mL of a human anti-TNF alpha antibody, or antigen-binding portion thereof.
  • the human antibody, or antigen-binding portion thereof comprises a light chain comprising a CDR3 domain comprising an amino acid sequence set forth as SEQ ID NO: 3 and a heavy chain comprising a CDR3 domain comprising an amino acid sequence set forth as SEQ ID NO: 4.
  • the antibody has a light chain CDR3 domain comprising the amino acid sequence of SEQ ID NO: 3, or modified from SEQ ID NO: 3 by a single alanine substitution at position 1, 4, 5, 7 or 8 or by one to five conservative amino acid substitutions at positions 1, 3, 4, 6, 7, 8 and/or 9 and has a heavy chain CDR3 domain comprising the amino acid sequence of SEQ ID NO: 4, or modified from SEQ ID NO: 4 by a single alanine substitution at position 2, 3, 4, 5, 6, 8, 9, 10 or 11 or by one to five conservative amino acid substitutions at positions 2, 3, 4, 5, 6, 8, 9, 10, 11 and/or 12.
  • the antibody of the invention may have certain functional characteristics.
  • the human antibody, or an antigen-binding portion thereof may dissociate from human TNF ⁇ with a K d of 1 ⁇ 10 ⁇ 8 M or less, dissociate from human TNF ⁇ with a K off rate constant of 1 ⁇ 10 ⁇ 3 s ⁇ 1 or less, both determined by surface plasmon resonance, and/or neutralize human TNF ⁇ cytotoxicity in a standard in vitro L929 assay with an IC 50 of 1 ⁇ 10 ⁇ 7 M or less.
  • the human antibody, or antigen-binding portion thereof is a human IgG1 kappa antibody.
  • the light chain of the human antibody, or antigen-binding portion thereof further comprises a CDR2 domain comprising an amino acid sequence set forth as SEQ ID NO: 5 and a CDR1 domain comprising an amino acid sequence set forth as SEQ ID NO: 7, and/or the heavy chain of the human antibody comprises a CDR2 domain comprising an amino acid sequence set forth as SEQ ID NO: 6 and a CDR1 domain comprising an amino acid sequence set forth as SEQ ID NO: 8.
  • the light chain of the human antibody, or antigen-binding portion thereof comprises the amino acid sequence set forth as SEQ ID NO: 1 and the heavy chain of the human antibody comprises the amino acid sequence set forth as SEQ ID NO: 2.
  • human antibodies or antigen-binding portions thereof, having amino acid sequences which are at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the SEQ ID NOs recited herein.
  • the human antibody, or antigen-binding portion thereof is adalimumab.
  • FIG. 1 is a graph depicting the presence of high molecular weight (hmw) protein specimen in a solution containing 0.1% Solutol. According to MALS (grey line), aggregate molar masses equal up to nearly 10 9 g/mol, accounting for 2.6% of total protein (UV280, black line). Storage at 40° C. for 12 w.
  • hmw high molecular weight
  • FIGS. 2A and 2B are graphs depicting the early-stage detection of high molecular weight (hmw) aggregates emerging during 40° C. storage. Whereas no aggregates could be detected via UV280 (black curve), MALS (grey curve) unambiguously proved the presence of hmw specimen.
  • FIG. 3 is a graph depicting the turbidity vs. freeze/thaw cycles of formulations F1-F6.
  • FIG. 4 is a graph depicting the polydispersity index vs. freeze/thaw cycles of formulations F1-F6.
  • FIG. 5 is a graph depicting the aggregate levels by SEC vs. freeze/thaw cycles of formulations F1-F6.
  • FIG. 6 is a graph depicting Tm in ° C. by DSC of formulations F1-F6 at T0.
  • FIG. 7 is a graph depicting aggregate levels by SEC vs. stirring time of formulations F1-F6.
  • FIG. 8 is a graph depicting the comparison of turbidity values obtained in stability studies after 3 months storage of F2, F6 and F7 (3 representative batches 01032-0134).
  • FIG. 9 is a graph depicting the comparison of visible particle values by DAC score obtained in stability studies after 3 months storage of F2, F6 and F7 (3 representative batches 01032-0134).
  • FIG. 12 is a graph depicting the comparison of residual monomer content obtained in stability studies after 3 months storage of F2, F6 and F7 (3 representative batches 01032-0134).
  • FIG. 13 is a graph depicting the comparison of sum of lysine variants obtained in stability studies after 3 months storage of F2, F6 and F7 (3 representative batches 01032-0134).
  • FIG. 14 is a graph depicting the turbidity data comparing F2, F6 and F7 in terms of stability against stir stress at different stirring speeds after 24 hours.
  • FIG. 15 is a graph depicting the DLS data (Z-average values) comparing F2, F6 and F7 in terms of stability against stir stress at different stirring speeds after 24 hours.
  • FIG. 16 is a graph depicting turbidity data comparing F2, F6 and F7 in terms of stability against stress before and after several pump cycles.
  • FIG. 17 is a graph depicting DLS data (Z-average) comparing F2, F6 and F7 in terms of stability before and after several pump cycles.
  • FIG. 18 is a graph depicting SEC data (aggregate levels) comparing F2, F6 and F7 in terms of stability before and after several pump cycles.
  • FIG. 19 is a graph depicting the visual score of 100 mg/mL formulations filled using a peristaltic pump.
  • FIG. 20 is a graph depicting the visual score of 100 mg/mL formulations filled using a piston pump.
  • FIG. 21 is a graph depicting the turbidity of 100 mg/mL formulations filled using a peristaltic pump.
  • FIG. 22 is a graph depicting the turbidity of 100 mg/mL formulations filled using a piston pump.
  • FIG. 23 is a graph depicting the turbidity at T0 and after 4 weeks storage at 5° C. of formulations F8-F11.
  • FIG. 24 is a graph depicting the monomer content at T0 and after 4 weeks storage at 5° C. of formulations F8-F11.
  • FIG. 25 is a graph depicting the aggregate levels at T0 and after 4 weeks storage at 5° C. of formulations F8-F11.
  • FIG. 26 is a graph depicting the subvisible particle count at T0 and after 4 weeks storage at 5° C. of formulations F8-F11.
  • pharmaceutical formulation refers to preparations which are in such form as to permit the biological activity of the active ingredients to be unequivocally effective, and which contain no additional components which are significantly toxic to the subjects to which the formulation would be administered.
  • phrases “pharmaceutically acceptable carrier” is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administration to mammals.
  • the carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to or impacting safety of the patient.
  • “Pharmaceutically acceptable excipients” are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
  • excipient refers to an agent which may be added to a formulation to provide a desired consistency, e.g., altering the bulk properties, to improve stability, and/or to adjust osmolality.
  • excipients include, but are not limited to, sugars, polyols, amino acids, surfactants, and polymers.
  • a commonly used excipient is a polyol.
  • a “polyol” is a substance with multiple hydroxyl groups, and includes sugars (reducing and nonreducing sugars), sugar alcohols and sugar acids.
  • Preferred polyols herein have a molecular weight which is less than about 600 kD (e.g., in the range from about 120 to about 400 kD).
  • Non-limiting examples of polyols are fructose, mannose, maltose, lactose, arabinose, xylose, ribose, rhamnose, galactose, glucose, sucrose, trehalose, sorbose, melezitose, raffinose, mannitol, xylitol, erythritol, threitol, sorbitol, glycerol, L-gluconate and metallic salts thereof.
  • buffer refers to a buffered solution that resists changes in pH by the action of its acid-base conjugate components.
  • the buffers of this invention have a pH in the range from about 4 to about 8; preferably from about 4.5 to about 7; and most preferably has a pH in the range from about 5.0 to about 6.5.
  • Examples of buffers that will control the pH in this range include phosphate, acetate (e.g., sodium acetate), succinate (such as sodium succinate), gluconate, glutamate, histidine, citrate and other organic acid buffers.
  • a buffer suitable for use in the formulations of the invention is a citrate and phosphate buffer.
  • surfactant generally includes those agents which protect a protein in a formulation from air/solution interface-induced stresses and solution/surface induced-stresses.
  • a surfactant may protect the protein from aggregation.
  • Suitable surfactants may include, e.g., polysorbates, polyoxyethylene alkyl ethers such as Brij 35.RTM., or poloxamer such as Tween 20, Tween 80, or poloxamer 188.
  • Preferred detergents are poloxamers, e.g., Poloxamer 188, Poloxamer 407; polyoxyethylene alkyl ethers, e.g.,Brij 35.RTM., Cremophor A25, Sympatens ALM/230; and polysorbates/Tweens, e.g., Polysorbate 20, Polysorbate 80, Mirj, and Poloxamers, e.g., Poloxamer 188, and Tweens, e.g., Tween 20 and Tween 80.
  • a “stable” formulation is one in which the antibody therein essentially retains its physical stability and/or chemical stability and/or biological activity during the manufacturing process and/or upon storage.
  • Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. (1993) Adv. Drug Delivery Rev. 10: 29-90.
  • the stability of the protein is determined according to the percentage of monomer protein in the solution, with a low percentage of degraded (e.g., fragmented) and/or aggregated protein.
  • the formulation is stable at room temperature (about 30° C.) or at 40° C. for at least 1 month and/or stable at about 2-8° C. for at least 1 year or for at least 2 years.
  • the formulation is preferably stable following freezing (to, e.g., ⁇ 70° C.) and thawing of the formulation, hereinafter referred to as a “freeze/thaw cycle.”
  • An antibody “retains its physical stability” in a pharmaceutical formulation if it shows substantially no signs of, e.g., aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography.
  • Aggregation is a process whereby individual molecules or complexes associate covalently or non-covalently to form aggregates. Aggregation can proceed to the extent that a visible precipitate is formed.
  • Stability such as physical stability of a formulation
  • a sample's apparent attenuation of light absorbance, or optical density
  • Such a measurement of light attenuation relates to the turbidity of a formulation.
  • the turbidity of a formulation is partially an intrinsic property of the protein dissolved in solution and is commonly determined by nephelometry, and measured in Nephelometric Turbidity Units (NTU).
  • NTU Nephelometric Turbidity Units
  • the degree of turbidity e.g., as a function of the concentration of one or more of the components in the solution, e.g., protein and/or salt concentration, is also referred to as the “opalescence” or “opalescent appearance” of a formulation.
  • the degree of turbidity can be calculated by reference to a standard curve generated using suspensions of known turbidity. Reference standards for determining the degree of turbidity for pharmaceutical compositions can be based on the European Pharmacopeia criteria (European Pharmacopoeia, Fourth Ed., Directorate for the Quality of Medicine of the Council of Europe (EDQM), France).
  • a clear solution is defined as one with a turbidity less than or equal to a reference suspension which has a turbidity of approximately 3 according to European Pharmacopeia standards.
  • Nephelometric turbidity measurements can detect Rayleigh scatter, which typically changes linearly with concentration, in the absence of association or nonideality effects. Other methods for assessing physical stability are well-known in the art.
  • Chemical stability can be assessed by, e.g., detecting and quantifying chemically altered forms of the antibody.
  • Chemical alteration may involve size modification (e.g. clipping) which can be evaluated using size exclusion chromatography, SDS-PAGE and/or matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI/TOF MS), for example.
  • size modification e.g. clipping
  • MALDI/TOF MS matrix-assisted laser desorption ionization/time-of-flight mass spectrometry
  • Other types of chemical alteration include charge alteration (e.g. occurring as a result of deamidation or oxidation) which can be evaluated by ion-exchange chromatography, for example.
  • An antibody “retains its biological activity” in a pharmaceutical formulation if the antibody in a pharmaceutical formulation is biologically active for its intended purpose. For example, biological activity is retained if the biological activity of the antibody in the pharmaceutical formulation is within about 30%, about 20%, or about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared (e.g., as determined in an antigen binding assay).
  • a “therapeutically effective amount” or “effective amount” of an antibody refers to an amount effective in the prevention or treatment or alleviation of a symptom of a disorder for the treatment of which the antibody is effective.
  • a “disorder” is any condition that would benefit from treatment with the antibody. This includes chronic and acute disorders or diseases including those pathological conditions which predisposes the subject to the disorder in question.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intriacranial, intraarticular, intraspinal and intrasternal injection and infusion.
  • systemic administration means the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • human TNF-alpha (abbreviated herein as hTNF-alpha, TNF ⁇ , or simply hTNF), as used herein, is intended to refer to a human cytokine that exists as a 17 kD secreted form and a 26 kD membrane associated form, the biologically active form of which is composed of a trimer of noncovalently bound 17 kD molecules.
  • hTNF-alpha The structure of hTNF-alpha is described further in, for example, Pennica, D., et al. (1984) Nature 312:724-729; Davis, J. M., et al. (1987) Biochem 26:1322-1326; and Jones, E. Y., et al.
  • human TNF-alpha is intended to include recombinant human TNF-alpha (rhTNF-alpha), which can be prepared by standard recombinant expression methods or purchased commercially (R & D Systems, Catalog No. 210-TA, Minneapolis, Minn.).
  • antibody is intended to refer to immunoglobulin molecules comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Other naturally occurring antibodies of altered structure, such as, for example, camelid antibodies, are also included in this definition.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the formulation contains an antibody with CDR1, CDR2, and CDR3 sequences like those described in U.S. Pat. Nos. 6,090,382 and 6,258,562, each incorporated by reference herein.
  • CDR refers to the complementarity determining region within a antibody variable sequence.
  • CDR1, CDR2 and CDR3 are three CDRs in each of the variable regions of the heavy chain and the light chain, which are designated CDR1, CDR2 and CDR3, for each of the variable regions.
  • the exact boundaries of these CDRs have been defined differently according to different systems.
  • the system described by Kabat (Id.) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody, but also provides precise residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Chothia et al.
  • CDR boundary definitions may not strictly follow one of the herein described systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding.
  • the methods used herein may utilize CDRs defined according to any of these systems, although certain embodiments use Kabat or Chothia defined CDRs.
  • antibody portion refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hTNF-alpha). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′) 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR).
  • a Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH1 domains
  • a F(ab′) 2 fragment a bivalent fragment comprising two Fab fragments linked by
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody.
  • Other forms of single chain antibodies, such as diabodies are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123).
  • the formulation contains an antigen-binding portions described in U.S. Pat. Nos. 6,090,382 and 6,258,562, each incorporated by reference herein.
  • an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecules, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides.
  • immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol.
  • Antibody portions such as Fab and F(ab′) 2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies.
  • antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques, as described herein.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies used in the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further in Section II, below), antibodies isolated from a recombinant, combinatorial human antibody library (described further in Section III, below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • an “isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds hTNF-alpha is substantially free of antibodies that specifically bind antigens other than hTNF-alpha).
  • An isolated antibody that specifically binds hTNF-alpha may, however, have cross-reactivity to other antigens, such as TNF-alpha molecules from other species.
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • a “neutralizing antibody”, as used herein is intended to refer to an antibody whose binding to hTNF-alpha results in inhibition of the biological activity of hTNF-alpha.
  • This inhibition of the biological activity of hTNF-alpha can be assessed by measuring one or more indicators of hTNF-alpha biological activity, such as hTNF-alpha-induced cytotoxicity (either in vitro or in vivo), hTNF-alpha-induced cellular activation and hTNF-alpha binding to hTNF-alpha receptors.
  • hTNF-alpha biological activity can be assessed by one or more of several standard in vitro or in vivo assays known in the art, and described in U.S. Pat. Nos. 6,090,382 and 6,258,562, each incorporated by reference herein.
  • the ability of an antibody to neutralize hTNF-alpha activity is assessed by inhibition of hTNF-alpha-induced cytotoxicity of L929 cells.
  • the ability of an antibody to inhibit hTNF-alpha-induced expression of ELAM-1 on HUVEC, as a measure of hTNF-alpha-induced cellular activation can be assessed.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • BIAcore Phharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.
  • K on is intended to refer to the on rate constant for association of a binding protein (e.g., an antibody) to the antigen to form the, e.g., antibody/antigen complex as is known in the art.
  • a binding protein e.g., an antibody
  • K off is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
  • Kd is intended to refer to the dissociation constant of a particular antibody-antigen interaction and refers to the value obtained in a titration measurement at equilibrium, or by dividing the dissociation rate constant (k off ) by the association rate constant (k on ).
  • the present invention features liquid pharmaceutical formulations (e.g., antibody formulations) having improved properties as compared to art-recognized formulations.
  • the present invention is based on the surprising finding that by removing NaCl and adding more than 20 mg/mL of a polyol, e.g., a sugar alcohol, the concentration of a human TNF alpha antibody in a formulation can be increased to about 100 mg/mL.
  • a polyol e.g., a sugar alcohol
  • the concentration of a human TNF alpha antibody in a formulation can be increased to about 100 mg/mL.
  • the formulation of the invention is able to maintain solubility and stability of the protein, e.g., during manufacturing, storage, and/or repeated freeze/thaw processing steps or extended exposure to increased air-liquid interfaces.
  • the formulation of the invention maintains a low level of protein aggregation (i.e., less than 1%), despite having about 100 mg/mL of antibody.
  • the formulation of the invention also, surprisingly, maintain a low viscosity within ranges suitable for subcutaneous injection, despite having about 100 mg/mL of antibody.
  • the formulation of the invention e.g., high concentration TNF alpha antibody, maintains solubility, maintains a low viscosity suitable for subcutaneous injection, and maintains stability over a pH range of almost one, e.g., pH 5.2 to pH 6.0.
  • turbidity of the formulation is less than 100 NTU after a standard 48 hour stir-stress assay.
  • the high antibody formulation of the invention overcomes a number of known challenges for formulations, including stability, viscosity, turbidity, and physical degradation challenges.
  • a surprising feature of the formulation of the invention is that in the absence of NaCl, the overall viscosity of the formulation remains low (e.g., about 3.1-3.3 mPas*s, e.g., about 3.00, 3.05, 3.10, 3.15, 3.20, 3.25, 3.30, 3.35, or about 3.40 mPas*s), while the antibody concentration is high (e.g., 100 mg/mL or greater).
  • viscosity increases as the protein concentration increases (see Shire et al. (2004) J Pharm Sci 93:1390 for review).
  • the liquid pharmaceutical formulation of the invention provides a high antibody concentration (e.g., at least 100 mg/mL) with a viscosity suitable for subcutaneous administration, without the need for the addition of NaCl.
  • formulations of the invention include high concentrations of proteins such that the liquid formulation does not show significant opalescence, aggregation, or precipitation.
  • formulations of the invention include high concentrations of proteins such that are suitable for, e.g., subcutaneous administration without significant felt pain (e.g., as determined by a visual analog scale (VAS) score).
  • VAS visual analog scale
  • the formulations of the invention comprise a high protein concentration, including, for example, a protein concentration about 50 mg/mL or about 100 mg/mL of a human anti-TNF-alpha antibody or antigen-binding fragment thereof. Accordingly, as described in Example 1 below, in one aspect of the invention the liquid pharmaceutical formulation comprises a human anti-TNF alpha antibody concentration of about 50 mg/mL. As described in Examples 2-6 below, in another aspect of the invention the liquid pharmaceutical formulation comprises a human anti-TNF alpha antibody concentration of about 100 mg/mL. In yet another aspect of the invention the liquid pharmaceutical formulation comprises a human anti-TNF alpha antibody concentration of about 150 mg/mL.
  • the preferred embodiments of the invention are formulations comprising high protein concentrations, it is also contemplated that the formulations of the invention may comprise an antibody concentration between about 1 mg/mL and about 150 mg/mL or about 40 mg/mL-125 mg/mL.
  • Concentrations and ranges intermediate to the above recited concentrations are also intended to be part of this invention (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24,25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108,
  • the invention provides a liquid pharmaceutical composition
  • a liquid pharmaceutical composition comprising a polyol, a surfactant, and a buffer system, in amounts sufficient to formulate an antibody, e.g., adalimumab, for therapeutic use at a concentration of greater than about, for example, 100 mg/mL.
  • the liquid pharmaceutical compositions do not comprise NaCl.
  • the preferred formulations of the invention do not comprise NaCl, a small amount of NaCl may be present in the formulations, e.g., from about 0.01 mM to about 300 mM. In addition, any amount of NaCl intermediate to the recited values are intended to be included.
  • the invention provides a liquid pharmaceutical composition
  • a liquid pharmaceutical composition comprising a human anti-TNF-alpha antibody or antigen binding fragment thereof, (e.g., adalimumab), a polyol, without the addition of NaCl, in amounts sufficient to formulate an antibody for therapeutic use.
  • a human anti-TNF-alpha antibody or antigen binding fragment thereof e.g., adalimumab
  • a polyol without the addition of NaCl
  • the present invention also provides liquid formulations comprising a human anti-TNF-alpha antibody or antigen binding fragment thereof, at a pH of about 5.0 to 6.4, and a turbidity of less than about 60 NTU after a standard 24 hour stir-stress assay, without the addition of NaCl (e.g., about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37. 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, or 63 NTU).
  • NaCl e.g., about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37. 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61,
  • the invention provides liquid formulations comprising a human anti-TNF-alpha antibody or antigen binding fragment thereof, at a pH of about 5.0 to 6.4, and a turbidity of less than about 100 NTU after a standard 48 hour stir-stress assay, without the addition of NaCl (e.g., about 35, 36, 37.
  • the invention provides liquid formulations comprising a human anti-TNF-alpha antibody or antigen binding fragment thereof, at a pH of about 5.0 to 6.4, and a turbidity of less than about 40 NTU after 3 months storage at 5° C., 25° C., or 40° C., without the addition of NaCl (e.g., about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37. 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60 NTU).
  • NaCl e.g., about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37. 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60 NTU.
  • a feature of the formulation of the invention is the inclusion of a polyol, e.g, a sugar alcohol, at a concentration of greater than 20 mg/mL.
  • the polyol is either sorbitol or mannitol. It should be noted that the addition of sorbitol or mannitol to protein solutions is not always associated with a gain in protein stability. For instance, sorbitol offered no advantage against precipitation of porcine growth hormone when evaluated during thermal or interfacial stress conditions—in contrast to Tween 20 and hydroxypropyl- ⁇ -cyclodextrin, respectively (Charman et al. (1993) Pharm Res.10(7):954-62).
  • a suitable polyol for use in the formulations of the invention is a sugar alcohol, e.g., mannitol or sorbitol.
  • the liquid formulations of the invention comprising a polyol typically comprise more than about 20 mg of the polyol. In one embodiment, the formulations comprise more than about 30 mg/mL of the polyol. In another embodiment, the formulations comprise more than about 40 mg/mL of the polyol. In another embodiment, he formulations comprise about 40-45 mg/mL of the polyol, e.g., about 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, or 55 mg/mL. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.
  • a liquid formulation comprising the antibody in a pH-buffered solution.
  • the buffer of this invention has a pH ranging from about 4 to about 8, preferably from about 4.5 to about 7.0, more preferably from about 4.5 to about 6.0, even more preferably from about 4.8 to about 5.5, and most preferably has a pH of about 5.0 to about 6.4.
  • the pH of the formulation of the invention is about 5.2.
  • the pH of the formulation of the invention is about 6.0.
  • Ranges intermediate to the above recited pH's are also intended to be part of this invention (e.g., 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4). Ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included, e.g., 5.2-5.8.
  • buffers that will control the pH within this range include phosphate, acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, glutamate, histidine, citrate and other organic acid buffers.
  • the formulation comprises a buffer system which contains citrate and/or phosphate to maintain the pH in a range of about 5.0 to about 6.4.
  • the pH of the formulation is about 5.2. In another embodiment, the pH of the formulation is about 6.0.
  • the buffer system includes citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, and/or sodium dihydrogen phosphate dihydrate.
  • the buffer system includes about 1.15-1.45 mg/ml of citric acid (e.g., about 1.15, 1.20, 1.25, 1.30, 1.35, 1.40, or 1.45), about 0.2-0.4 mg/mL of sodium citrate dehydrate (e.g., about 0.2, 0.25, 0.3, 0.35, or 0.4), about 1.35-1.75 mg/mL of disodium phosphate dehydrate (e.g., 1.35, 1.40, 1.45, 1.50, 1.55, 1.60, 1.65, 1.70, or 1.75), about 0.75-0.95 mg/mL of sodium dihydrogen phosphate dehydrate (e.g., about 0.75, 0.80, 0.85, 0.9, or 0.95).
  • the buffer system includes 1.3-1.31 mg/mL of citric acid (e.g., about 1.305 mg/mL). In another embodiment, the buffer system includes about 0.27-0.33 mg/mL of sodium citrate dehydrate (e.g., about 0.305 mg/mL). In one embodiment, the buffer system includes about 1.5-1.56 mg/mL of disodium phosphate dehydrate (e.g., about 1.53 mg/mL). In another embodiment, the buffer system includes about 0.83-0.89 mg/mL of sodium dihydrogen phosphate dihydrate (e.g., about 0.86 mg/mL).
  • a detergent or surfactant may also be added to the antibody formulation of the invention.
  • exemplary detergents include nonionic detergents such as polysorbates (e.g. polysorbates 20, 80, etc.) or poloxamers (e.g. poloxamer 188).
  • the amount of detergent added is such that it reduces aggregation of the formulated antibody and/or minimizes the formation of particulates in the formulation and/or reduces adsorption.
  • the formulation includes a surfactant which is a polysorbate.
  • the formulation contains the detergent polysorbate 80.
  • the formulation contains between about 0.1 and about 2.0 mg/mL of polysorbate 80, e.g., about 1 mg/mL.
  • the formulation of the invention consists essentially of a human TNF alpha antibody, or antigen binding portion thereof, at a concentration of at least about 100 mg/mL, a surfactant (e.g., polysorbate 80), a polyol (e.g., more than 20 mg/mL of sorbitol or mannitol), and a buffering system (e.g., citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, and/or sodium dihydrogen phosphate dihydrate), and does not contain NaCl.
  • a surfactant e.g., polysorbate 80
  • a polyol e.g., more than 20 mg/mL of sorbitol or mannitol
  • a buffering system e.g., citric acid monohydrate, sodium citrate, disodium phosphate dihydrate, and/or sodium dihydrogen phosphate dihydrate
  • the formulation contains the above-identified agents (i.e., an antibody at a concentration of at least about 100 mg/mL, a buffer system, a polyol, and a surfactant, without NaCl) and is essentially free of preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl.
  • a preservative may be included in the formulation.
  • One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980) may be included in the formulation provided that they do not significantly adversely affect the desired characteristics of the formulation.
  • Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include; additional buffering agents; co-solvents; antioxidants including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium.
  • the formulation herein may also be combined with one or more other therapeutic agents as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect the antibody of the formulation.
  • Such therapeutic agents are suitably present in combination in amounts that are effective for the purpose intended. Additional therapeutic agents which can be combined with the formulation of the invention are further described in U.S. Pat. Nos. 6,090,382 and 6,258,562, each of which is incorporated herein by reference.
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes prior to, or following, preparation of the formulation.
  • the liquid formulation of the invention has advantageous stability and storage properties. Stability of the liquid formulation is not dependent on the form of storage, and includes, but is not limited to, formulations which are frozen, lyophilized, spray-dried, or formulations which in which the active ingredient is suspended. Stability can be measured at a selected temperature for a selected time period.
  • the protein in the liquid formulations is stable in a liquid form for at least about 3 months; at least about 4 months, at least about 5 months; at least about 6 months; at least about 12 months; at least about 18 months.
  • the formulation is stable at room temperature (about 30° C.) or at 40° C. for at least about 1 month and/or stable at about 2-8° C. for at least about 1 year, or more preferably stable at about 2-8° C. for at least about 2 years.
  • the formulation is preferably stable following freezing (to, e.g., ⁇ 80° C.) and thawing of the formulation, hereinafter referred to as a “freeze/thaw cycle.”
  • Stability of a protein in a liquid formulation may also be defined as the percentage of monomer, aggregate, or fragment, or combinations thereof, of the protein in the formulation.
  • a protein “retains its physical stability” in a formulation if it shows substantially no signs of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography.
  • a stable liquid formulation is a formulation having less than about 10%, and preferably less than about 5% of the protein being present as aggregate in the formulation.
  • the physical stability of a liquid formulation is determined by determining turbidity of the formulation following a stir stress assay, e.g., 24 hour or 48-hour stir-stress assay.
  • a stir stress assay may be performed by placing a suitable volume of a liquid formulation in a beaker with a magnetic stirrer, e.g., (multipoint HP, 550 rpm), removing aliquots at any suitable time, e.g., at T0-T48 (hrs), and performing suitable assays as desired on the aliquots. Samples of a formulation under the same conditions but without stirring serve as control.
  • Turbidity measurements may be performed using a laboratory turbidity measurement system from Hach (Germany) and are reported as nephelometric units (NTU).
  • the liquid formulations of the invention also have advantageous tolerability properties. Tolerability is evaluated based on assessment of subject-perceived injection site pain using the Pain Visual Analog Scale (VAS).
  • VAS Pain Visual Analog Scale
  • VAS is a measurement instrument that measures pain as it ranges across a continuum of values, e.g., from none to an extreme amount of pain.
  • a VAS is a horizontal line, about 100 mm in length, anchored by numerical and/or word descriptors, e.g., 0 or 10, or ‘no pain’ or ‘excruciating pain’, optionally with additional word or numeric descriptors between the extremes, e.g., mild, moderate, and severe; or 1 through 9) (see, e.g., Lee J S, et al. (2000) Acad Emerg Med 7:550).
  • Draize Scale hereinafter “hemorrhage, petechiae, erythema, edema, pruritus
  • bruising e.g., the Draize Scale (hemorrhage, petechiae, erythema, edema, pruritus) and bruising.
  • Antibodies that can be used in the formulations of the invention are antibodies directed against the antigen TNF-alpha, including human TNF-alpha (or hTNF-alpha).
  • the invention features an isolated human antibody, or antigen-binding portion thereof, that binds to human TNF-alpha with high affinity and a low off rate, and also has a high neutralizing capacity.
  • the human antibodies used in the invention are recombinant, neutralizing human anti-hTNF-alpha antibodies.
  • the most preferred recombinant, neutralizing antibody of the invention is referred to herein as D2E7, also referred to as HUMIRATM or adalimumab (the amino acid sequence of the D2E7 VL region is shown in SEQ ID NO: 1; the amino acid sequence of the D2E7 VH region is shown in SEQ ID NO: 2).
  • D2E7 (adalimumab/HUMIRA®) have been described in Salfeld et al., U.S. Pat. Nos. 6,090,382, 6,258,562, and 6,509,015, which are each incorporated by reference herein.
  • the human TNF-alpha, or an antigen-binding portion thereof dissociates from human TNF-alpha with a Kd of 1 ⁇ 10-8 M or less and a Koff rate constant of 1 ⁇ 10-3 s-1 or less, both determined by surface plasmon resonance, and neutralizes human TNF-alpha cytotoxicity in a standard in vitro L929 assay with an IC50 of 1 ⁇ 10-7 M or less. More preferably, the isolated human antibody, or antigen-binding portion thereof, dissociates from human TNF-alpha with a Koff of 5 ⁇ 10-4 s-1 or less, or even more preferably, with a Koff of 1 ⁇ 10-4 s-1 or less.
  • the isolated human antibody, or antigen-binding portion thereof neutralizes human TNF-alpha cytotoxicity in a standard in vitro L929 assay with an IC50 of 1 ⁇ 10-8 M or less, even more preferably with an IC50 of 1 ⁇ 10-9 M or less and still more preferably with an IC50 of 1 ⁇ 10-10 M or less.
  • the antibody is an isolated human recombinant antibody, or an antigen-binding portion thereof.
  • the invention pertains to treating Crohn's disease by administering human antibodies that have slow dissociation kinetics for association with hTNF-alpha and that have light and heavy chain CDR3 domains that structurally are identical to or related to those of D2E7.
  • Position 9 of the D2E7 VL CDR3 can be occupied by Ala or Thr without substantially affecting the Koff.
  • a consensus motif for the D2E7 VL CDR3 comprises the amino acid sequence: Q-R-Y-N-R-A-P-Y-(T/A) (SEQ ID NO: 3).
  • a consensus motif for the D2E7 VH CDR3 comprises the amino acid sequence: V-S-Y-L-S-T-A-S-S-L-D-(Y/N) (SEQ ID NO: 4).
  • the CDR3 domain of the D2E7 heavy and light chains is amenable to substitution with a single alanine residue (at position 1, 4, 5, 7 or 8 within the VL CDR3 or at position 2, 3, 4, 5, 6, 8, 9, 10 or 11 within the VH CDR3) without substantially affecting the Koff.
  • substitutions by alanine substitution of other amino acids within the CDR3 domains may be possible while still retaining the low off rate constant of the antibody, in particular substitutions with conservative amino acids.
  • no more than one to five conservative amino acid substitutions are made within the D2E7 VL and/or VH CDR3 domains.
  • no more than one to three conservative amino acid substitutions are made within the D2E7 VL and/or VH CDR3 domains.
  • conservative amino acid substitutions should not be made at amino acid positions critical for binding to hTNF alpha.
  • Positions 2 and 5 of the D2E7 VL CDR3 and positions 1 and 7 of the D2E7 VH CDR3 appear to be critical for interaction with hTNF alpha and thus, conservative amino acid substitutions preferably are not made at these positions (although an alanine substitution at position 5 of the D2E7 VL CDR3 is acceptable, as described above) (see U.S. Pat. No. 6,090,382).
  • the antibody or antigen-binding portion thereof preferably contains the following characteristics:
  • a) dissociates from human TNF ⁇ with a Koff rate constant of 1 ⁇ 10-3 s-1 or less, as determined by surface plasmon resonance;
  • b) has a light chain CDR3 domain comprising the amino acid sequence of SEQ ID NO: 3, or modified from SEQ ID NO: 3 by a single alanine substitution at position 1, 4, 5, 7 or 8 or by one to five conservative amino acid substitutions at positions 1, 3, 4, 6, 7, 8 and/or 9;
  • c) has a heavy chain CDR3 domain comprising the amino acid sequence of SEQ ID NO: 4, or modified from SEQ ID NO: 4 by a single alanine substitution at position 2, 3, 4, 5, 6, 8, 9, 10 or 11 or by one to five conservative amino acid substitutions at positions 2, 3, 4, 5, 6, 8, 9, 10, 11 and/or 12.
  • the antibody, or antigen-binding portion thereof dissociates from human TNF-alpha with a Koff of 5 x 10-4 s-1 or less. Even more preferably, the antibody, or antigen-binding portion thereof, dissociates from human TNF-alpha with a Koff of 1 ⁇ 10-4 s-1 or less.
  • the antibody or antigen-binding portion thereof preferably contains a light chain variable region (LCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 3, or modified from SEQ ID NO: 3 by a single alanine substitution at position 1, 4, 5, 7 or 8, and with a heavy chain variable region (HCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 4, or modified from SEQ ID NO: 4 by a single alanine substitution at position 2, 3, 4, 5, 6, 8, 9, 10 or 11.
  • LCVR light chain variable region
  • HCVR heavy chain variable region
  • the LCVR further has a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 5 (i.e., the D2E7 VL CDR2) and the HCVR further has a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 6 (i.e., the D2E7 VH CDR2).
  • the LCVR further has CDR1 domain comprising the amino acid sequence of SEQ ID NO: 7 (i.e., the D2E7 VL CDR1) and the HCVR has a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 8 (i.e., the D2E7 VH CDR1).
  • the framework regions for VL preferably are from the V ⁇ I human germline family, more preferably from the A20 human germline Vk gene and most preferably from the D2E7 VL framework sequences shown in FIGS. 1A and 1B of U.S. Pat. No. 6,090,382.
  • the framework regions for VH preferably are from the VH3 human germline family, more preferably from the DP-31 human germline VH gene and most preferably from the D2E7 VH framework sequences shown in FIGS. 2A and 2B of U.S. Pat. No. 6,090,382.
  • the antibody or antigen-binding portion thereof preferably contains a light chain variable region (LCVR) comprising the amino acid sequence of SEQ ID NO: 1 (i.e., the D2E7 VL) and a heavy chain variable region (HCVR) comprising the amino acid sequence of SEQ ID NO: 2 (i.e., the D2E7 VH).
  • the antibody comprises a heavy chain constant region, such as an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region.
  • the heavy chain constant region is an IgG1 heavy chain constant region or an IgG4 heavy chain constant region.
  • the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region.
  • the antibody comprises a kappa light chain constant region.
  • the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
  • the invention includes uses of an isolated human antibody, or an antigen-binding portion thereof, containing D2E7-related VL and VH CDR3 domains.
  • a light chain variable region having a CDR3 domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25 and SEQ ID NO: 26 or with a heavy chain variable region (HCVR) having a CDR3 domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30,
  • An antibody, or antibody portion, used in the methods and compositions of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell.
  • a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed in the host cell and, preferably, secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered.
  • Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel, F. M. et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Pat. No. 4,816,397 by Boss et al.
  • DNA fragments encoding the light and heavy chain variable regions are first obtained. These DNAs can be obtained by amplification and modification of germline light and heavy chain variable sequences using the polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • Germline DNA sequences for human heavy and light chain variable region genes are known in the art (see e.g., the “Vbase” human germline sequence database; see also Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Tomlinson, I.
  • a member of the VH3 family of human germline VH genes is amplified by standard PCR.
  • the DP-31 VH germline sequence is amplified.
  • a member of the V ⁇ I family of human germline VL genes is amplified by standard PCR.
  • the A20 VL germline sequence is amplified.
  • these sequences can be mutated to encode the D2E7 or D2E7-related amino acid sequences disclosed herein.
  • the amino acid sequences encoded by the germline VH and VL DNA sequences are first compared to the D2E7 or D2E7-related VH and VL amino acid sequences to identify amino acid residues in the D2E7 or D2E7-related sequence that differ from germline. Then, the appropriate nucleotides of the germline DNA sequences are mutated such that the mutated germline sequence encodes the D2E7 or D2E7-related amino acid sequence, using the genetic code to determine which nucleotide changes should be made.
  • Mutagenesis of the germline sequences is carried out by standard methods, such as PCR-mediated mutagenesis (in which the mutated nucleotides are incorporated into the PCR primers such that the PCR product contains the mutations) or site-directed mutagenesis.
  • the “germline” sequences obtained by PCR amplification encode amino acid differences in the framework regions from the true germline configuration (i.e., differences in the amplified sequence as compared to the true germline sequence, for example as a result of somatic mutation), it may be desirable to change these amino acid differences back to the true germline sequences (i.e., “backmutation” of framework residues to the germline configuration).
  • DNA fragments encoding D2E7 or D2E7-related VH and VL segments are obtained (e.g., by amplification and mutagenesis of germline VH and VL genes, as described above), these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to convert the variable region genes to full-length antibody chain genes, to Fab fragment genes or to a scFv gene.
  • a VL- or VH-encoding DNA fragment is operatively linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker.
  • the term “operatively linked”, as used in this context, is intended to mean that the two DNA fragments are joined such that the amino acid sequences encoded by the two DNA fragments remain in-frame.
  • the isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CH1, CH2 and CH3).
  • heavy chain constant regions CH1, CH2 and CH3
  • the sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the heavy chain constant region can be an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an IgG1 or IgG4 constant region.
  • the VH-encoding DNA can be operatively linked to another DNA molecule encoding only the heavy chain CH1 constant region.
  • the isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL.
  • the sequences of human light chain constant region genes are known in the art (see e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the light chain constant region can be a kappa or lambda constant region, but most preferably is a kappa constant region.
  • the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser)3, such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242:423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al., Nature (1990) 348:552-554).
  • a flexible linker e.g., encoding the amino acid sequence (Gly4-Ser)3
  • DNAs encoding partial or full-length light and heavy chains, obtained as described above, are inserted into expression vectors such that the genes are operatively linked to transcriptional and translational control sequences.
  • operatively linked is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into separate vector or, more typically, both genes are inserted into the same expression vector.
  • the antibody genes are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • the expression vector Prior to insertion of the D2E7 or D2E7-related light or heavy chain sequences, the expression vector may already carry antibody constant region sequences.
  • one approach to converting the D2E7 or D2E7-related VH and VL sequences to full-length antibody genes is to insert them into expression vectors already encoding heavy chain constant and light chain constant regions, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector and the VL segment is operatively linked to the CL segment within the vector.
  • the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell.
  • the antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene.
  • the signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the antibody chain genes in a host cell.
  • the term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes.
  • Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • Preferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • the recombinant expression vectors used in the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes.
  • the selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al.).
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • DHFR dihydrofolate reductase
  • the expression vector(s) encoding the heavy and light chains is transfected into a host cell by standard techniques.
  • the various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R. J. Kaufman and P. A. Sharp (1982) Mol. Biol. 159:601-621), NSO myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr- CHO cells described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R. J. Kaufman and P. A. Sharp (1982) Mol. Biol. 159:601-621
  • NSO myeloma cells
  • the antibodies When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
  • Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or all of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to hTNF alpha. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
  • bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than hTNF alpha by cros slinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection.
  • the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes.
  • the recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification.
  • the selected transformant host cells are culture to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium.
  • Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • the nucleotide sequence encoding the D2E7 light chain variable region is shown in SEQ ID NO: 36.
  • the CDR1 domain of the LCVR encompasses nucleotides 70-102, the CDR2 domain encompasses nucleotides 148-168 and the CDR3 domain encompasses nucleotides 265-291.
  • nucleotide sequence encoding the D2E7 heavy chain variable region is shown in SEQ ID NO: 37.
  • the CDR1 domain of the HCVR encompasses nucleotides 91-105
  • the CDR2 domain encompasses nucleotides 148-198
  • the CDR3 domain encompasses nucleotides 295-330.
  • nucleotide sequences encoding D2E7-related antibodies, or portions thereof e.g., a CDR domain, such as a CDR3 domain
  • the liquid pharmaceutical formulation comprises a human TNF alpha antibody, or antigen-binding portion thereof, that is a bioequivalent or biosimilar to the antibody adalimumab.
  • a biosimilar antibody is an antibody which shows no clinically meaningful difference when compared to a reference antibody, e.g., adalimumab.
  • a biosimilar antibody has equivalent safety, purity, and potency as a reference antibody, e.g., adalimumab.
  • An advantage of the formulation of the invention is that is may be used to deliver a high concentration of a human anti-TNF alpha antibody, or antigen-binding portion, (e.g., adalimumab) to a subject subcutaneously.
  • a human anti-TNF alpha antibody, or antigen-binding portion e.g., adalimumab
  • the formulation of the invention are delivered to a subject subcutaneously.
  • the subject administers the formulation to himself/herself.
  • an effective amount of the formulation is administered.
  • the language “effective amount” of the formulation is that amount necessary or sufficient to inhibit TNF-alpha activity, e.g., prevent the various morphological and somatic symptoms of a detrimental TNF-alpha activity-associated state.
  • the effective amount of the formulation is the amount necessary to achieve the desired result.
  • An example of an effective amount of the formulation is an amount sufficient to inhibit detrimental TNF-alpha activity or treat a disorder in which TNF alpha activity is detrimental.
  • a disorder in which TNF-alpha activity is detrimental is intended to include diseases and other disorders in which the presence of TNF-alpha. in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which TNF-alpha. activity is detrimental is a disorder in which inhibition of TNF-alpha. activity is expected to alleviate the symptoms and/or progression of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of TNF-alpha. in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of TNF-alpha. in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an anti-TNF-alpha. antibody.
  • an increase in the concentration of TNF-alpha. in a biological fluid of a subject suffering from the disorder e.g., an increase in the concentration of
  • one advantage of the formulations of the invention is the ability to prepare formulations comprising high concentrations of antibody without increasing the viscosity of the formualtion. Therefore, as also described below, the new formulations permit administration of high amounts (e.g., effective amounts) of antibody in smaller volumes as compared to prior commercial formulations, thereby decreasing pain.
  • the effective amount of antibody may be determined according to a strictly weight based dosing scheme (e.g., mg/kg) or may be a total body dose (also referred to as a fixed dose) which is independent of weight.
  • an effective amount of the formulation is 0.8 mL of the formulation containing a total body dose of about 80 mg of antibody (i.e., 0.8 mL of a 100 mg/mL antibody formulation of the invention).
  • an effective amount of the formulation is 0.4 mL of the formulation of the invention containing a total body dose of about 40 mg of antibody (i.e., 0.4 mL of a 100 mg/mL antibody formulation of the invention).
  • an effective amount of the formulation is twice 0.8 mL of the formulation containing a total body dose of about 160 mg of antibody (i.e., two units containing 0.8 mL each of a 100 mg/mL antibody formulation of the invention).
  • an effective amount of the formulation is 0.2 mL of the formulation of the invention containing a total body dose of about 20 mg of antibody (i.e., 0.2 mL of a 100 mg/mL antibody formulation of the invention).
  • an effective amount may be determined according to a weight-based fixed dosing regimen (see, e.g., WO 2008/154543, incorporated by reference herein).
  • the invention provides a stable, high concentration formulation with an extended shelf life, which, in one embodiment, is used to inhibit TNF-alpha activity in a subject suffering from a disorder in which TNF-alpha activity is detrimental, comprising administering to the subject a formulation of the invention such that TNF-alpha activity in the subject is inhibited.
  • the TNF-alpha is human TNF-alpha and the subject is a human subject.
  • the subject can be a mammal expressing a TNF-alpha with which an antibody of the invention cross-reacts.
  • the subject can be a mammal into which has been introduced hTNF-alpha (e.g., by administration of hTNF-alpha or by expression of an hTNF-alpha transgene).
  • a formulation of the invention can be administered to a human subject for therapeutic purposes (discussed further below).
  • the liquid pharmaceutical formulation is easily administratable, which includes, for example, a formulation which is self-administered by the patient.
  • the formulation of the invention is administered through subcutaneous injection, preferably single use.
  • a formulation of the invention can be administered to a non-human mammal expressing a TNF-alpha with which the antibody cross-reacts (e.g., a primate, pig or mouse) for veterinary purposes or as an animal model of human disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of antibodies of the invention (e.g., testing of dosages and time courses of administration).
  • the liquid pharmaceutical formulation of the invention may be administered to a subject via a prefilled syringe, an autoinjector pen, or a needle-free administration device.
  • the invention also features an autoinjector pen, a prefilled syringe, or a needle-free administration device comprising the liquid pharmaceutical formulation of the invention.
  • the invention features a delivery device comprising a dose of the formulation comprising 100 mg/mL a human TNF alpha antibody, or antigen-binding portion thereof, e.g., an autoinjector pen or prefilled syringe comprises a dose of about 19 mg, 20, mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 51 mg, 52 mg, 53 mg, 54 mg, 55 mg, 56 mg, 57 mg, 58 mg, 59 mg, 60 mg, 61 mg, 62 mg, 63 mg, 64 mg, 65 mg, 66 mg, 67 mg, 68 mg, 69 mg, 70 mg, 71 mg, 72 mg, 73 mg, 74 mg, 75 mg,
  • the formulation of the invention is used to treat disorders in which TNF alpha activity is detrimental.
  • a disorder in which TNF-alpha activity is detrimental is intended to include diseases and other disorders in which the presence of TNF-alpha in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which TNF-alpha activity is detrimental is a disorder in which inhibition of TNF-alpha activity is expected to alleviate the symptoms and/or progression of the disorder.
  • Such disorders may be evidenced, for example, by an increase in the concentration of TNF-alpha in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of TNF-alpha in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an anti-TNF-alpha antibody as described above.
  • a biological fluid of a subject suffering from the disorder e.g., an increase in the concentration of TNF-alpha in serum, plasma, synovial fluid, etc. of the subject
  • an anti-TNF-alpha antibody as described above.
  • TNF-alpha activity is detrimental.
  • TNF-alpha activity is detrimental are also described in U.S. Pat. Nos. 6,015,557; 6,177,077; 6,379,666; 6,419,934; 6,419,944; 6,423,321; 6,428,787; and 6,537,549; and PCT Publication Nos. WO 00/50079 and WO 01/49321, the entire contents of all of which are incorporated herein by reference.
  • the formulations of the invention may also be used to treat disorders in which TNF alpha activity is detrimental as described in U.S. Pat. Nos. 6,090,382, 6,258,562 and U.S. Patent Application Publication No. US20040126372, the entire contents of all of which are incorporated herein by reference.
  • Tumor necrosis factor has an established role in the pathophysiology of sepsis, with biological effects that include hypotension, myocardial suppression, vascular leakage syndrome, organ necrosis, stimulation of the release of toxic secondary mediators and activation of the clotting cascade (see e.g., Tracey, K. J. and Cerami, A. (1994) Annu. Rev. Med. 45:491-503; Russell, D and Thompson, R. C. (1993) Curr. Opin. Biotech. 4:714-721). Accordingly, the formulation of the invention can be used to treat sepsis in any of its clinical settings, including septic shock, endotoxic shock, gram negative sepsis and toxic shock syndrome.
  • the formulation of the invention can be coadministered with one or more additional therapeutic agents that may further alleviate sepsis, such as an interleukin-1 inhibitor (such as those described in PCT Publication Nos. WO 92/16221 and WO 92/17583), the cytokine interleukin-6 (see e.g., PCT Publication No. WO 93/11793) or an antagonist of platelet activating factor (see e.g., European Patent Application Publication No. EP 374 510).
  • an interleukin-1 inhibitor such as those described in PCT Publication Nos. WO 92/16221 and WO 92/17583
  • the cytokine interleukin-6 see e.g., PCT Publication No. WO 93/11793
  • an antagonist of platelet activating factor see e.g., European Patent Application Publication No. EP 374 510.
  • the formulation of the invention is administered to a human subject within a subgroup of sepsis patients having a serum or plasma concentration of IL-6 above 500 pg/ml, and more preferably 1000 pg/ml, at the time of treatment (see PCT Publication No. WO 95/20978).
  • Tumor necrosis factor has been implicated in playing a role in the pathophysiology of a variety of autoimmune diseases.
  • TNF-alpha has been implicated in activating tissue inflammation and causing joint destruction in rheumatoid arthritis (see e.g., Tracey and Cerami, supra; Arend, W. P. and Dayer, J-M. (1995) Arth. Rheum. 38:151-160; Fava, R. A., et al. (1993) Clin. Exp. Immunol. 94:261-266).
  • TNF-alpha also has been implicated in promoting the death of islet cells and in mediating insulin resistance in diabetes (see e.g., Tracey and Cerami, supra; PCT Publication No.
  • TNF-alpha also has been implicated in mediating cytotoxicity to oligodendrocytes and induction of inflammatory plaques in multiple sclerosis (see e.g., Tracey and Cerami, supra).
  • JIA juvenile idiopathic arthritis
  • JIA also referred to as juvenile rheumatoid arthritis
  • Grom et al. (1996) Arthritis Rheum. 39:1703; Mangge et al. (1995) Arthritis Rheum. 8:211).
  • the formulation of the invention can be used to treat autoimmune diseases, in particular those associated with inflammation, including rheumatoid arthritis, rheumatoid spondylitis (also referred to as ankylosing spondylitis), osteoarthritis and gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis, juvenile idiopathic arthritis (also referred to as juvenile rheumatoid arthritis), and nephrotic syndrome.
  • autoimmune diseases in particular those associated with inflammation, including rheumatoid arthritis, rheumatoid spondylitis (also referred to as ankylosing spondylitis), osteoarthritis and gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis, juvenile idiopathic arthritis (also referred to as juvenile rheumatoid arthritis), and nephrotic syndrome.
  • Tumor necrosis factor has been implicated in mediating biological effects observed in a variety of infectious diseases.
  • TNF-alpha has been implicated in mediating brain inflammation and capillary thrombosis and infarction in malaria (see e.g., Tracey and Cerami, supra).
  • TNF-alpha also has been implicated in mediating brain inflammation, inducing breakdown of the blood-brain barrier, triggering septic shock syndrome and activating venous infarction in meningitis (see e.g., Tracey and Cerami, supra).
  • TNF-alpha also has been implicated in inducing cachexia, stimulating viral proliferation and mediating central nervous system injury in acquired immune deficiency syndrome (AIDS) (see e.g., Tracey and Cerami, supra).
  • infectious diseases including bacterial meningitis (see e.g., European Patent Application Publication No. EP 585 705), cerebral malaria, AIDS and AIDS-related complex (ARC) (see e.g., European Patent Application Publication No. EP 230 574), as well as cytomegalovirus infection secondary to transplantation (see e.g., Fietze, E., et al. (1994) Transplantation 58:675-680).
  • the formulation of the invention also can be used to alleviate symptoms associated with infectious diseases, including fever and myalgias due to infection (such as influenza) and cachexia secondary to infection (e.g., secondary to AIDS or ARC).
  • Tumor necrosis factor has been implicated as a key mediator of allograft rejection and graft versus host disease (GVHD) and in mediating an adverse reaction that has been observed when the rat antibody OKT3, directed against the T cell receptor CD3 complex, is used to inhibit rejection of renal transplants (see e.g., Tracey and Cerami, supra; Eason, J. D., et al. (1995) Transplantation 59:300-305; Suthanthiran, M. and Strom, T. B. (1994) New Engl. J. Med. 331:365-375). Accordingly, the formulations of the invention can be used to inhibit transplant rejection, including rejections of allografts and xenografts and to inhibit GVHD.
  • GVHD graft versus host disease
  • the antibody or antibody portion may be used alone, it can be used in combination with one or more other agents that inhibit the immune response against the allograft or inhibit GVHD.
  • the formulations of the invention are used in combination with OKT3 to inhibit OKT3-induced reactions.
  • the formulation of the invention is used in combination with one or more antibodies directed at other targets involved in regulating immune responses, such as the cell surface molecules CD25 (interleukin-2 receptor-.alpha.), CD11a (LFA-1), CD54 (ICAM-1), CD4, CD45, CD28/CTLA4, CD80 (B7-1) and/or CD86 (B7-2).
  • the formulation of the invention is used in combination with one or more general immunosuppressive agents, such as cyclosporin A or FK506.
  • Tumor necrosis factor has been implicated in inducing cachexia, stimulating tumor growth, enhancing metastatic potential and mediating cytotoxicity in malignancies (see e.g., Tracey and Cerami, supra). Accordingly, the formulations of the invention can be used in the treatment of malignancies, to inhibit tumor growth or metastasis and/or to alleviate cachexia secondary to malignancy.
  • Tumor necrosis factor has been implicated in the pathophysiology of adult respiratory distress syndrome, including stimulating leukocyte-endothelial activation, directing cytotoxicity to pneumocytes and inducing vascular leakage syndrome (see e.g., Tracey and Cerami, supra). Accordingly, the formulations of the invention can be used to treat various pulmonary disorders, including adult respiratory distress syndrome (see e.g., PCT Publication No. WO 91/04054), shock lung, chronic pulmonary inflammatory disease, pulmonary sarcoidosis, pulmonary fibrosis and silicosis.
  • Tumor necrosis factor has been implicated in the pathophysiology of inflammatory bowel disorders (see e.g., Tracy, K. J., et al. (1986) Science 234:470-474; Sun, X-M., et al. (1988) J. Clin. Invest. 81:1328-1331; MacDonald, T. T., et al. (1990) Clin. Exp. Immunol. 81:301-305) Chimeric murine anti-hTNF-alpha antibodies have undergone clinical testing for treatment of Crohn's disease (van Dullemen, H. M., et al. (1995) Gastroenterology 109:129-135).
  • the formulation of the invention also can be used to treat intestinal disorders, such as idiopathic inflammatory bowel disease, which includes two syndromes, Crohn's disease and ulcerative colitis.
  • the formulation of the invention also can be used to treat various cardiac disorders, including ischemia of the heart (see e.g., European Patent Application Publication No. EP 453 898) and heart insufficiency (weakness of the heart muscle)(see e.g., PCT Publication No. WO 94/20139).
  • ischemia of the heart see e.g., European Patent Application Publication No. EP 453 898
  • heart insufficiency weakness of the heart muscle
  • TNF ⁇ has been implicated in the pathophysiology of a wide variety of disorders, including inflammatory diseases such as spondyloarthopathies (see e.g., Moeller, A., et al. (1990) Cytokine 2:162-169; U.S. Pat. No. 5,231,024 to Moeller et al.; European Patent Publication No. 260 610 B1 by Moeller, A).
  • An example of a spondyloarthropathy that may be treated by the formulation of the invention includes psoriatic arthritis.
  • Tumor necrosis factor has been implicated in the pathophysiology of psoriatic arthritis (Partsch et al. (1998) Ann Rheum Dis. 57:691; Ritchlin et al. (1998) J Rheumatol. 25:1544).
  • the formulation of the invention is used to treat skin and nail disorders.
  • skin and nail disorder in which TNF ⁇ activity is detrimental is intended to include skin and/or nail disorders and other disorders in which the presence of TNF alpha in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder, e.g., psoriasis.
  • An example of a skin disorder which may be treated using the formulation of the invention is psoriasis.
  • the formulation of the invention is used to treat plaque psoriasis.
  • Tumor necrosis factor has been implicated in the pathophysiology of psoriasis (Takematsu et al. (1989) Arch Dermatol Res. 281:398; Victor and Gottling (2002) J Drugs Dermatol. 1(3):264).
  • the formulation of the invention is used to treat rheumatoid arthritis, psoriatic arthritis, or ankylosing spondylitis.
  • the formulation of the invention comprising an isolated human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab), may be administered to a human subject according to a dosing scheme and dose amount effective for treating rheumatoid arthritis, psoriatic arthritis, or ankylosing spondylitis.
  • a dose of about 40 mg of a human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab) (e.g., 0.4 mL of a 100 mg/mL formulation of the invention) in the formulation of the invention is administered to a human subject every other week for the treatment of rheumatoid arthritis, psoriatic arthritis, or ankylosing spondylitis.
  • a human TNF alpha antibody, or antigen-binding portion thereof, e.g., adalimumab
  • a dose of about 40 mg of a human TNF alpha antibody, or antigen-binding portion thereof, in the formulation of the invention is administered to a human subject every other week for the treatment of rheumatoid arthritis, psoriatic arthritis, or ankylosing spondylitis.
  • the formulation is administered subcutaneously, every other week (also referred to as biweekly, see methods of administration described in US20030235585, incorporated by reference herein) for the treatment of rheumatoid arthritis, ankylosing spondylitis, or psoriatic arthritis.
  • the formulation of the invention is used to treat Crohn's disease.
  • the formulation of the invention comprising an isolated human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab), may be administered to a human subject according to a dosing scheme and dose amount effective for treating Crohn's disease.
  • a dose of about 160 mg of a human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab) (e.g., 1.6 mL of a 100 mg/mL formulation of the invention) in the formulation of the invention is administered to a human subject initially at about day 1, followed by a subsequent dose of 80 mg of the antibody (e.g., 0.8 mL of a 100 mg/mL formulation of the invention) two weeks later, followed by administration of about 40 mg (e.g., 0.4 mL of a 100 mg/mL formulation of the invention) every other week for the treatment of Crohn's disease.
  • adalimumab e.g., 1.6 mL of a 100 mg/mL formulation of the invention
  • 80 mg of the antibody e.g., 0.8 mL of a 100 mg/mL formulation of the invention
  • administration of about 40 mg e.g., 0.4 mL of a 100 mg/
  • the formulation is administered subcutaneously, according to a multiple variable dose regimen comprising an induction dose(s) and maintenance dose(s) (see, for example, U.S. Patent Publication Nos. US20060009385 and US20090317399) for the treatment of Crohn's disease, each of which are incorporated by reference herein) for the treatment of Crohn's disease.
  • the formulation of the invention is used to treat psoriasis.
  • the formulation of the invention comprising an isolated human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab), may be administered to a human subject according to a dosing scheme and dose amount effective for treating psoriasis.
  • an initial dose of about 80 mg of a human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab) (e.g., 0.8 mL of a 100 mg/mL formulation of the invention) in the formulation of the invention is administered to a human subject, followed by a subsequent dose of 40 mg of the antibody (e.g., 0.4 mL of a 100 mg/mL formulation of the invention) every other week starting one week after the initial dose.
  • a human TNF alpha antibody, or antigen-binding portion thereof, e.g., adalimumab
  • a subsequent dose of 40 mg of the antibody e.g., 0.4 mL of a 100 mg/mL formulation of the invention
  • the formulation is administered subcutaneously, according to a multiple variable dose regimen comprising an induction dose(s) and maintenance dose(s) (see, for example, US 20060009385 and WO 2007/120823, each of which are incorporated by reference herein) for the treatment of psoriasis.
  • the formulation of the invention is used to treat juvenile idiopathic arthritis (JIA).
  • the formulation of the invention comprising an isolated human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab), may be administered to a human subject according to a dosing scheme and dose amount effective for treating JIA.
  • 20 mg of a human TNF alpha antibody, or antigen-binding portion thereof, in the formulation of the invention e.g., 0.2 mL of a 100 mg/mL formulation of the invention
  • 40 mg of a human TNF alpha antibody, or antigen-binding portion thereof, in the formulation of the invention is administered to a subject weighing more than or equal to 30 kg (66 lbs) every other week for the treatment of JIA.
  • the formulation is administered subcutaneously, according to a weight-based fixed dose (see, for example, U.S. Patent Publication No. 20090271164, incorporated by reference herein) for the treatment of JIA.
  • an isolated human TNF alpha antibody, or antigen-binding portion thereof, may be administered to a human subject for treatment of a disorder associated with detrimental TNF ⁇ activity according to a monthly dosing schedule, whereby the antibody is administered once every month or once every four weeks.
  • disorders that may be treated according to a monthly dosing schedule include, but are not limited to, rheumatoid arthritis, ankylosing spondylitis, JIA, psoriasis, Crohn's disease, or psoriatic arthritis.
  • the formulation of the invention comprising an isolated human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab), may be administered to a human subject for treatment of a disorder associated with detrimental TNF ⁇ activity according to a monthly dosing schedule.
  • 80 mg of a human TNF alpha antibody, or antigen-binding portion thereof, in the formulation of the invention e.g., 0.8 mL of a 100 mg/mL formulation of the invention
  • a subject having a disorder associated with detrimental TNF ⁇ activity is administered to a subject having a disorder associated with detrimental TNF ⁇ activity.
  • Dose amounts described herein may be delivered as a single dose (e.g., a single dose of 40 mg in 0.4 mL or 80 mg dose in 0.8 mL), or, alternatively may be delivered as multiple doses (e.g., four 40 mg doses or two 80 mg doses for delivery of a 160 mg dose).
  • the formulation of the invention comprising an isolated human TNF alpha antibody, or antigen-binding portion thereof, (e.g., adalimumab) may also be administered to a subject in combination with an additional therapeutic agent.
  • the formulation is administered to a human subject for treatment of rheumatoid arthritis in combination with methotrexate or other disease-modifying anti-rheumatic drugs (DMARDs).
  • DMARDs disease-modifying anti-rheumatic drugs
  • the formulation is administered to a human subject for treatment of JIA in combination with methotrexate or other disease-modifying anti-rheumatic drugs (DMARDs). Additional combination therapies are described in U.S. Pat. Nos. 6,258,562 and 7,541,031; and U.S. Patent Publication No. US20040126372, the entire contents of all of which are incorporated by reference herein.
  • the formulation of the invention comprising a human TNF alpha antibody, or antigen-binding portion thereof, may also be used to treat a subject who has failed previous TNF inhibitor therapy, e.g., a subject who has lost response to or is intolerant to infliximab.
  • This Example provides results of experiments aimed at improving the stability of the pharmaceutical formulation of the antibody adalimumab.
  • Adalimumab (subclass G 1 , about 47 kDa) was formulated in a modified pharmaceutical formulation in order to generate a liquid parenteral dosage form at 50 mg/mL final drug concentration.
  • Previous formulation experiments had determined that a phosphate/citrate buffer system was superior to other buffer systems in terms of protein stabilization of adalimumab. Consequently, improved stability was addressed via addition of excipients for a liquid 50 mg/mL dosage. All excipients used were of highest purity (“pro analysis” grade) and purchased from Merck KGaA, Darmstadt, Germany. Mannitol was sourced from Mallinckrodt Baker B. V., Deventer, Holland.
  • adalimumab concentrates ⁇ 70 mg/mL
  • excipient stock solutions ⁇ 70 mg/mL
  • the 70 mg/mL adalimumab stock solution was prepared using a composition of citrate and phosphate buffer components (i.e., citric acid * H 2 O, sodium citrate dehydrate, Na 2 HPO 4 * 2H 2 O, NaH 2 PO 4 * 2H 2 O) as listed in Table 16.
  • citric acid * H 2 O sodium citrate dehydrate
  • Na 2 HPO 4 * 2H 2 O NaH 2 PO 4 * 2H 2 O
  • Excipient stock solutions were generated by excipient dissolution in phosphate/citrate buffer medium using a composition of citrate and phosphate buffer components (i.e., citric acid * H2O, sodium citrate dehydrate, Na 2 HPO 4 * 2H 2 O, NaH 2 PO 4 * 2H 2 O) as listed in Table 16.
  • citric acid * H2O sodium citrate dehydrate
  • Na 2 HPO 4 * 2H 2 O NaH 2 PO 4 * 2H 2 O
  • Table 16 Prior to sterile filtration (0.2 ⁇ m, Minisart®, Sartorius AG, Goettingen, Germany), pH adjustment was performed by adding of acid/base specimen of buffer components.
  • the various formulations were subjected to 3 month-short-time storage at three different temperatures (5° C., 25° C., 40° C.).
  • Adalimumab concentrates were provided by diafiltration of adalimumab bulk solution via Vivaflow 50 units (cut-off 50 kDa, Vivascience G, Hannover, Germany), using phosphate/citrate buffer medium for buffer exchange.
  • Current processes for concentration and buffer exchange of biopharmaceutical solutions are based on IEX, SE-HPLC, ultra-/diafiltration and tangential flow filtration (Christy et al. (2002) Desalination, 144:133-136). Diafiltration was applied because purification, concentration and buffer exchange are possible within a single-unit operation with variable flow dynamics, thus minimizing protein stress (Table 1).
  • adalimumab concentrates were centrifugated (5° C., 3000 g, 20 minutes).
  • Tween 80 was proposed as an ingredient that could alleviate particle contamination in solutions containing salts, such as NaCl (Tables 4A and 4B). Solutions were then examined that contained both 6.16 mg/mL NaCl and 1 mg/mL Tween 80.
  • Tween 80 In order to determine the stabilizing potential of surfactants on 50 mg/mL adalimumab formulation, various amounts of Tween 80 (0.%, 0.03%, 0.1%) were added to a protein solution containing 6.16 mg/mL NaCl. Generally, Tween 80 is assumed to stabilize proteins e.g., by binding through hydrophobic surface interaction. As a protein's surface characteristics are influenced by the presence of salts, the effect of the absence of NaCl additionally was surveyed (described as 0.1% Tween 80 solution without NaCl in Table 5) (see also Kheirolomoom et al. (1998) Biochem. Eng. J., 2:81-88).
  • Tween 80 The results from varying amounts of Tween 80 with and without NaCal are presented in Table 5. As shown, Tween 80 was unable to provide stability to the formulation with or without NaCl. With respect to 0.03% Tween 80/NaCl, the combination resulted in decreasing the monomer content after 12 weeks of storage at 40° C. This result contradicted the majority of articles addressing this topic, as generally the stabilizing impact of Tween 80 is related to increasing concentrations of surfactant (valid in the range from 0.001 to 1%) (see Arakawa et al. (2001) Adv. Drug Deliv. Rev., 46:307-326).
  • Tween 80 proved to confer notable stability towards adalimumab during freeze-thaw cycles (Table 7).
  • Tween 80 was also determined by repeatedly subjecting the solutions to stress via freezing ( ⁇ 80° C., 12 hours) and thawing (5° C., 12 hours). The number of freeze-thaw (freeze/thaw) cycles applied was closely correlated to a gain in subvisible particulate matter. However, whereas the effect of 5 freeze/thaw cycles on solutions with 0 or 0.03% Tween 80 content resulted in a ⁇ 10-fold increase in particle contamination (particles ⁇ 1 ⁇ m), the situation virtually remained unchanged in 0.1% Tween 80 solutions. SE-HPLC analysis confirmed these results (Table 8).
  • the nonionic surfactant Solutol® HS15 was investigated for its potential to stabilize adalimumab.
  • the protecting features of Solutol® in concentrations of 0.03 and 0.1% were shown recently in terms of expcumin parenterals (Steckel et al. (2003) Int. J. Pharm., 257:181-194).
  • the influence of Solutol® on adalimumab solutions in terms of the formation of particulate matter contamination were compared to protein solutions containing 0.1% Tween 80 (Table 9).
  • Subvisible Subvisible Subvisible Particles Particles Particles Particles Subvisible Particles >1 ⁇ m/mL Content >1 ⁇ m/mL Content >1 ⁇ m/mL Content >1 ⁇ m/mL Content Storage Temp.
  • sugars e.g., sucrose, glucose, raffinose, trehalose
  • polyols e.g., glycerol, sorbitol, mannitol
  • polyols e.g., glycerol, sorbitol, mannitol
  • polyols such as sorbitol are often used to stabilize parenterals, for instance in a number of lyophilized vaccine pharmaceuticals such as MumpsvaxTM, MeruvaxTM II and AttenuvaxTM or intravenous administrable solutions such as CardeneTM.
  • sugars and polyols In contrast to other excipients such as surfactants, sugars and polyols must be added in higher concentrations (>0.5 M) in order to deploy their complete stabilizing potential. As a consequence, sorbitol at concentrations of 50 and 100 mg/mL was added to adalimumab solutions, and subjected to 12 weeks of storage (Table 10).
  • Sorbitol decreased the tendency for particle formation during storage, compared to solutions where no sorbitol was present. The amount of added sorbitol did virtually not result in any differences. Regarding monomer content, the stabilizing effect of sorbitol was found to be closely concentration-dependent. The presence of NaCl detracts from protein stability (Table 11).
  • adalimumab at a concentration of 50 mg/mL was effectively stabilized by adding mannitol or sorbitol to the formulation. Besides contributing to protein stability by native state protection, mannitol and sorbitol stabilized the protein via a further mechanism, thereby reducing fragmentation during long-term storage.
  • NaCl is the most-used salt in the formulation of protein parenterals. Nevertheless, the above results show that, at an adalimumab concentration of 50 mg/mL, NaCl impeded adalimumab stability in the presence of polyols, and did not increase protein stability as a sole excipient.
  • consideration of their behaviour in accordance with the Hoffmeister lyotropic series provided a rough rule of thumb.
  • anionic acetate instead of chloride as counterion in sodium salts was investigated.
  • the individual solutions i.e., 50 mg/mL sorbitol/4 mg/mL Na-acetate, 50 mg/mL sorbitol/4 mg/mL NaCal, and 50 mg/mL sorbitol, no salt
  • the adalimumab solution containing NaCl was stacked against protein stability, since after only 4 weeks of storage (40° C.) a comparison of formulations containing either NaCl or sodium acetate showed that the monomer content in the sodium acetate enriched batch was ⁇ 0.25% greater than that of the NaCl containing formulation, adding up to a >0.4% difference after 12 weeks. Consequently, sodium acetate contributed more to adalimumab stability than sodium chloride. Nevertheless, the addition of sodium acetate did not increase protein stabilization, since the salt-free formulation had identical monomer content.
  • acetate containing formulations In comparison to both other formulations (formulations with 50 mg/mL Sorbitol and wither no salt of 4 mg/mL NaCl), acetate containing formulations exhibited a greater number of particles beyond 1 ⁇ m (180,000/mL versus ⁇ 6,000/mL).
  • Buffer systems were also examined, whereby sodium and potassium buffer systems were compared with varying concentrations of sorbitol. As illustrated in Table 15, the stability of adalimumab dissolved in potassium phosphate buffer equaled that determined in sodium phosphate buffers. Data of storage tests performed at 25° C. substantiated these findings. Additionally, both buffer systems equaled in particulate matter contamination. Thus, potassium phosphate was considered to be preferred in liquid protein formulations.
  • adalimumab solutions at 50 mg/mL. If the presence of salts is favored, e.g., by reasons of osmolality—the sodium acetate has advantages over sodium chloride. Similarly, potassium based phosphate buffer systems equaled sodium phosphate buffer systems in terms of adalimumab stability.
  • the following example provides the ingredients for a number of high concentration protein formulations comprising the ant-TNF ⁇ antibody adalimumab. Surprisingly, the formulations described below had a number of advantageous properties, despite the high concentration of antibody, i.e., about 100 mg/mL.
  • F1 to F6 A number of characteristics of the formulations (referred to as F1 to F6) were studied relative to the commercial 50 mg/mL adalimumab formulation (F7), including turbidity.
  • the turbidity of the solutions was determined by analysis of the undiluted solution. Turbidity is reported as NTU values (Nephelometric Turbidity Units).
  • Visible particle contamination was determined by visual inspection as described in German Drug Codex. Subvisible particles were monitored by the light obscuration method according to USP. Dynamic light scattering analysis of diluted solutions was employed to assess the hydrodynamic diameter (reported as the mean or Z-average size calculated by cumulants analysis of the DLS measured intensity autocorrelation function and polydispersity index, PDI, of the size distribution of particles).
  • CEX-HPLC Cation Exchange HPLC
  • the formulations tested are referenced as F1-F6 (Table 16), containing 100 mg/mL adalimumab in different matrices spanning from pH 5.2 to pH 6.0, formulated with different polyols and with or without sodium chloride.
  • Adalimumab 100 100 100 100 100 100 50 Mannitol 12 42 — 12 42 — 12 Sorbitol — — 42 — — 42 — Polysorbate 80 1 1 1 1 1 1 1 1 1 citric acid * H 2 O 1.305 1.305 1.305 1.305 1.305 1.305 1.305 Sodium citrate 0.305 0.305 0.305 0.305 0.305 0.305 0.305 0.305 dehydrate Na 2 HPO 4 * 2 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53 1.53
  • the starting solution is a solution of purified antibody at low concentration (lower than the high concentrations of the invention) in a liquid buffer, for example in a buffer resulting from the preceding manufacturing process step.
  • adalimumab solution was provided at a concentration of about 70 mg/mL in a buffer system identical to F7 without surfactant at pH 5.2.
  • the starting solution is then concentrated and diafiltered by ultrafiltration, preferably in a tangential-flow filtration system, using a membrane able to retain quantitatively the antibody, for example with a cutoff of 10 kD.
  • the representative formulations F2 and F6 were manufactured by diluting the concentrate to about 50 mg/L using the corresponding matrix without surfactant as diafiltration buffer.
  • a continuous buffer exchange was conducted using the tangential-flow filtration system.
  • the diafiltration was generally carried out at constant retentate volume, with at least 5 volumes, or preferably 8 volumes, of diafiltration buffer.
  • the diafiltered solution was further concentrated to a high concentration, for example higher or equal to 150 mg/mL.
  • the final turbid retentate was then recovered out of the ultrafiltration system by flushing the tubes with diafiltration buffer.
  • freeze/thaw stress (freezing performed at ⁇ 80° C., thawing performed at 25° C.) experiments were carried out.
  • Turbidity was measured as an indicator of the development of particle aggregates in the colloidal or in the visible range.
  • the turbidity (reported as NTU values) did not change significantly even after the fourth cycle of freeze/thaw ( FIG. 3 ).
  • Increased turbidity of solutions of higher pH may be attributed to increased protein-protein interactions due to lowered charge repulsion at the pH approaching the pI of the protein (adalimumab 8.5) (Wang et al. (2007) J Pharm Sci 96 (1) 2457-2468).
  • Dynamic light scattering was employed as a method for determining particle size in the submicron range.
  • the polydispersity index value obtained in the course of the size distribution determination was used as another sensitive indicator of aggregation in the colloidal or in the micrometer size range. Similar to the turbidity data, none of the tested formulations showed any signs of physical instability ( FIG. 4 ).
  • FIG. 5 depicts aggregate levels. No signs of physico-chemical instabilities were detected in relation to the repeated freeze/thawing stress.
  • DSC Differential Scanning Calorimetry
  • FIG. 6 provides Tm values for the 100 mg/mL adalimumab formulations. These data showed that all formulations achieve high Tm values. However, the sodium chloride free formulations (F2, F3, F5, F6) showed significantly increased Tm values indicating the robustness of these formulations. Since formulations are tested at 1 mg/mL, the Tm data of F1 is the same as the Tm of F7, thereby confirming the improved stability of the 100 mg/mL formulations without sodium chloride or at pH 6.0 over the F7 formulation.
  • a stir stress model using magnetic stir bars was used to detect physico-chemical instabilities of the new adalimumab formulations.
  • This well known model induces stress by subjecting adalimumab to long term air-liquid interface exposition as well as stirring related cavitation which leads to formation of soluble and insoluble protein aggregates in a predictable manner.
  • proteins formulated at pH values in the range of their respective pI are more susceptible to air-liquid interface related aggregation due to reduced repulsive forces.
  • ionic excipients such as sodium chloride, play a role in protein aggregation due to their ionic shielding properties. Hydrophobic attractive forces may be reduced with the presence of sodium chloride thereby reducing protein-protein interactions and increasing the colloidal stability (Shire et al. (2004) J Pharm Sci, 93 (6)1390-1402).
  • Turbidity data were evaluated to detect aggregate formation induced by stir stress.
  • Table 17 depicts nephelometric values in relation to the formulation composition and stirring time.
  • Initial turbidity values for F1-F3 (formulated at lower pH of 5.2) demonstrated differences between sodium chloride containing (F1) and NaCl free (F2, F3) solutions.
  • solutions adjusted to a higher pH of 6.0 (F4-F6) were characterized by higher turbidity. It is known in the art that NaCl may reduce the clarity of mAb solutions after mechanical stress such as stirring (e.g., Fesinmeyer et al. (2009) Pharm Res, 26 (4)903-913).
  • AN opalescent appearance is a simple consequence of Rayleigh scatter and linearly related to protein concentration.
  • opalescent appearance does not result in physical instability (Sukumar et al. (2004) Pharm Res 21 (7)1087-1093).
  • the 50 mg/mL adalimumab formulation showed turbidity of 63-130 NTU after 24 hours stirring and 109-243 NTU after 48 hours, whereas the 100 mg/mL formulations of adalimumab resulted in values ranging between 27-63 (24 hours) and 40-87 (48 hours). According to Treuheit et al.
  • the new formulations have increased stability compared to the 50 mg/mL formulation.
  • the new 100 mg/mL adalimumab formulations were subjected to long term storage to verify superior stability compared to the 50 mg/mL standard formulation. Stability data over 12 months at 5° C. (recommended storage temperature for the commercial product) were evaluated. The data indeed suggest that the new formulations displayed no reduced stability (Table 19).
  • particle levels did not change significantly throughout the 12 months stability testing and remained at significantly lower levels than F7.
  • Turbidity data from these batches verifies the superior behavior of the NaCl free formulations at 100 mg/mL, especially at the lower pH of 5.2.
  • Increasing the concentration of protein in solution is generally known to increase opalescence and thereby the turbidity readout due to Rayleigh scattering (Sukumar et al. (2004) Pharm Res 21 (7)1087-1093).
  • the new formulations without sodium chloride revealed similar turbidity levels at the same pH of the 50 mg/mL formulations ( FIG. 8 ).
  • FIGS. 9-11 provide detailed data of particulate formation (visible and subvisible particles) of the new formulations. The surprising finding of increased stability was verified. In fact, it was possible to reduce the both subvisible and visible particle score even after 3 months storage at elevated temperature.
  • Viscosity values were determined initially as a basic parameter characterizing the processability of protein solutions. Table 20 provides viscosity data obtained for the F1-F7 formulations. Increasing protein concentration led to increased viscosities compared to the 50 mg/mL formulation (F7).
  • Removal of the electrostatically shielding agent NaCl is expected to increase hydrophobic protein interactions, especially at pH values approaching the pI of adalimumab, thereby increasing the viscosity. This effect was reported to be most pronounced at NaCl concentration ⁇ 200 mM (Shire et al. (2004) J Pharm Sci, 93 (6)1390-1402).
  • peristaltic filling did not result in visible particle generation directly after filling (T0) and after storage.
  • piston filling resulted in significant particle counts even at T0 for the solutions formulated at pH 6.0 ( FIG. 20 ).
  • the highest values were measured in F4, containing sodium chloride, whereas F5-F6 resulted in significant lower scores, verifying the improved stability of sodium chloride free formulations against process stress.
  • Mannitol or sorbitol was used at a concentration of 42 mg/mL to meet tonicity requirements of sodium chloride-free solutions. Data showed that in comparison to a formerly used concentration of 12 mg/mL, both polyols not only contributed to the osmolality of the solutions, but additionally had a significant impact on protein stability.
  • Stability data suggested improved clarity for higher polyol concentrations, independent of the type of the polyol. Under conditions that are generally rated as not optimal (e.g., pH 6.0 close to the pI of adalimumab), formulations with higher polyol concentrations showed improved clarity even after short storage of 4 weeks at 5° C. This was observed with several analytical methods.
  • Adalimumab formulations were tested for the suitability to maintain Adalimumab physical and chemical stability under both accelerated stability test conditions and long-term storage at recommended storage temperature conditions (see Table 1 below).
  • Formulations differed in pH (pH 5.2 vs. pH 6), excipient conditions (e.g., concentrations of mannitol or sorbitol), salt/ionic strength conditions (e.g., concentration of NaCl), and protein concentration (50 mg/mL vs. 100 mg/mL).
  • Formulations F2 and F6 were identified as formulations that maintain both the physical and chemical stability of Adalimumab for at least 18 months and 12 months, respectively.
  • An exchange of the formulation excipient NaCl with mannitol (formulation F2) and sorbitol (formulation F6) conveys high stabilization potential, despite a 100% increase in protein concentration (from 50 mg/mL in formulation F7 to 100 mg/mL in formulations F2 and F6).
  • physical stability in both formulations were maintained for at least 12 and 18 months, respectively. Even after 12 months storage, both formulations contained more than 99% monomer (SEC data), and aggregate levels were below 1%.
  • the flinching model was chosen to assess injection site pain, and was used to evaluate impact of formulation composition on tolerability and pain sensations.
  • the new 100 mg/mL formulations were optimized to reduce subcutaneous injection-related side effects such as pain at the injection site.
  • Injection site pain comprises both pain related to the needle prick and sensations related to the infusion of the solution into the subQ depot.
  • certain needle designs may be advantageous to reduce injection site discomfort, no clear data on the formulation contribution was available (see, e.g., Chan, G. C. F., et al. (2003) American Journal of Hematology 76(4):398-404).
  • test formulation F7 Number of paw flinches 0-10 min post injection for active injections
  • Formulations tested were F2 (at pH 5.2), F5, and F7, the corresponding formulations at pH values closer to the physiological conditions.
US12/772,595 2009-05-04 2010-05-03 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies Abandoned US20100278822A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/772,595 US20100278822A1 (en) 2009-05-04 2010-05-03 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US14/170,061 US20140141008A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US14/170,026 US20140141007A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US17538009P 2009-05-04 2009-05-04
US12/772,595 US20100278822A1 (en) 2009-05-04 2010-05-03 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/170,026 Continuation US20140141007A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US14/170,061 Continuation US20140141008A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies

Publications (1)

Publication Number Publication Date
US20100278822A1 true US20100278822A1 (en) 2010-11-04

Family

ID=43030509

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/772,595 Abandoned US20100278822A1 (en) 2009-05-04 2010-05-03 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US14/170,026 Abandoned US20140141007A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US14/170,061 Abandoned US20140141008A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/170,026 Abandoned US20140141007A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US14/170,061 Abandoned US20140141008A1 (en) 2009-05-04 2014-01-31 Stable high protein concentration formulations of human anti-tnf-alpha-antibodies

Country Status (16)

Country Link
US (3) US20100278822A1 (es)
EP (1) EP2427211A4 (es)
JP (1) JP2012526121A (es)
KR (1) KR20120038406A (es)
CN (2) CN102458469B (es)
AR (1) AR076748A1 (es)
AU (1) AU2010246168A1 (es)
CA (1) CA2760185A1 (es)
IL (1) IL215643A0 (es)
MX (1) MX2011011772A (es)
NZ (2) NZ613809A (es)
RU (1) RU2560701C2 (es)
SG (2) SG10201401995UA (es)
TW (2) TWI480064B (es)
UY (1) UY32609A (es)
WO (1) WO2010129469A1 (es)

Cited By (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030235585A1 (en) * 2001-06-08 2003-12-25 Fischkoff Steven A. Methods of administering anti-TNFalpha antibodies
US20040126372A1 (en) * 2002-07-19 2004-07-01 Abbott Biotechnology Ltd. Treatment of TNFalpha related disorders
US20060083741A1 (en) * 2004-10-08 2006-04-20 Hoffman Rebecca S Treatment of respiratory syncytial virus (RSV) infection
US20060153846A1 (en) * 2002-08-16 2006-07-13 Hans-Juergen Krause Formulation of human antibodies for treating tnf-alpha associated disorders
US20080292642A1 (en) * 2007-03-29 2008-11-27 Borhani David W Crystalline anti-human IL-12 antibodies
US20080311043A1 (en) * 2006-06-08 2008-12-18 Hoffman Rebecca S Uses and compositions for treatment of psoriatic arthritis
US20090110679A1 (en) * 2007-07-13 2009-04-30 Luk-Chiu Li Methods and compositions for pulmonary administration of a TNFa inhibitor
US20090155205A1 (en) * 1996-02-09 2009-06-18 Salfeld Jochen G HUMAN ANTIBODIES THAT BIND HUMAN TNFa
US20090271164A1 (en) * 2008-01-03 2009-10-29 Peng Joanna Z Predicting long-term efficacy of a compound in the treatment of psoriasis
US20090280065A1 (en) * 2006-04-10 2009-11-12 Willian Mary K Uses and Compositions for Treatment of Psoriasis
US20090291062A1 (en) * 2007-11-30 2009-11-26 Wolfgang Fraunhofer Protein formulations and methods of making same
US20090304682A1 (en) * 2004-04-09 2009-12-10 Hoffman Rebecca S Multiple-variable dose regimen for treating TNFa-related disorders
US20090317399A1 (en) * 2006-04-10 2009-12-24 Pollack Paul F Uses and compositions for treatment of CROHN'S disease
US20100016557A1 (en) * 1996-02-09 2010-01-21 Abbott Biotechnology Ltd. HUMAN ANTIBODIES THAT BIND HUMAN TNFalpha
US20100021451A1 (en) * 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20100034823A1 (en) * 2006-10-27 2010-02-11 Borhani David W Crystalline anti-hTNFalpha antibodies
US20110002935A1 (en) * 2006-04-05 2011-01-06 Min Wan Antibody purification
US20110171227A1 (en) * 2006-04-10 2011-07-14 Okun Martin M Methods and compositions for treatment of skin disorders
WO2011097301A2 (en) 2010-02-02 2011-08-11 Abbott Biotechnology Ltd. METHODS AND COMPOSITIONS FOR PREDICTING RESPONSIVENESS TO TREATMENT WITH TNF-α INHIBITOR
US8162887B2 (en) 2004-06-23 2012-04-24 Abbott Biotechnology Ltd. Automatic injection devices
WO2012065072A3 (en) * 2010-11-11 2012-07-26 Abbott Biotechnology Ltd. IMPROVED HIGH CONCENTRATION ANTI-TNFα ANTIBODY LIQUID FORMULATIONS
WO2013096835A1 (en) * 2011-12-23 2013-06-27 Abbvie Inc. Stable protein formulations
US8636704B2 (en) 2009-04-29 2014-01-28 Abbvie Biotechnology Ltd Automatic injection device
US8658773B2 (en) * 2011-05-02 2014-02-25 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
US20140056888A1 (en) * 2002-09-06 2014-02-27 Alexion Pharmaceuticals, Inc. High concentration antibody formulations
WO2014039903A2 (en) 2012-09-07 2014-03-13 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US8679061B2 (en) 2006-06-30 2014-03-25 Abbvie Biotechnology Ltd Automatic injection device
US8715664B2 (en) 2005-05-16 2014-05-06 Abbvie Biotechnology Ltd. Use of human TNFα antibodies for treatment of erosive polyarthritis
US8747854B2 (en) 2010-06-03 2014-06-10 Abbvie Biotechnology Ltd. Methods of treating moderate to severe hidradenitis suppurativa with anti-TNF-alpha antibodies
US8753839B2 (en) 2007-08-08 2014-06-17 Abbvie Inc. Compositions and methods for crystallizing antibodies
US8758301B2 (en) 2009-12-15 2014-06-24 Abbvie Biotechnology Ltd Firing button for automatic injection device
WO2014099636A1 (en) * 2012-12-18 2014-06-26 Merck Sharp & Dohme Corp. Liquid formulations for an anti-tnf alpha antibody
WO2014114651A1 (en) * 2013-01-24 2014-07-31 Glaxosmithkline Intellectual Property Development Limited Tnf-alpha antigen-binding proteins
JP2014202667A (ja) * 2013-04-08 2014-10-27 株式会社島津製作所 抗体医薬用粒子径分布測定装置及び抗体医薬の粒子径分布測定方法
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
WO2015057910A1 (en) 2013-10-16 2015-04-23 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
CN104707146A (zh) * 2013-12-16 2015-06-17 浙江海正药业股份有限公司 一种含有阿达木单抗的药物组合物
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9086418B2 (en) 2005-11-01 2015-07-21 Abbvie Biotechnology Ltd. Methods and compositions for diagnosing ankylosing spondylitis using biomarkers
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US20150283241A1 (en) * 2012-10-26 2015-10-08 Lupin Limited Stable pharmaceutical composition of tnfr:fc fusion protein
US20150290325A1 (en) * 2012-11-19 2015-10-15 Merck Sharp & Dohme Corp. LIQUID FORMULATIONS FOR TNFR:Fc FUSION PROTEINS
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
EP2946767A1 (en) * 2014-05-23 2015-11-25 Ares Trading S.A. Liquid pharmaceutical composition
EP2946765A1 (en) * 2014-05-23 2015-11-25 Ares Trading S.A. Liquid pharmaceutical composition
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9279015B2 (en) 2006-04-10 2016-03-08 Robert L. Wong Methods for treatment of ankylosing spondylitis using TNF alpha antibodies
WO2016057739A1 (en) * 2014-10-09 2016-04-14 Regeneron Pharmaceuticals, Inc. Process for reducing subvisible particles in a pharmaceutical formulation
EP3009450A1 (en) * 2011-07-19 2016-04-20 Glaxo Group Limited Liquid formulation comprising adalimumab and an acetate buffer
US20160129112A1 (en) * 2013-05-28 2016-05-12 Momenta Pharmaceuticals, Inc. Pharmaceutical Compositions Comprising Pyrophosphate
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
WO2016120413A1 (en) * 2015-01-28 2016-08-04 Mabxience S.A. Pharmaceutical formulations for anti-tnf-alpha antibodies
EP3053573A1 (en) * 2015-02-06 2016-08-10 Ares Trading S.A. Liquid pharmaceutical composition
EP3078675A1 (en) 2015-04-10 2016-10-12 Ares Trading S.A. Induction dosing regimen for the treatment of tnf alpha mediated disorders
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9610301B2 (en) 2008-01-15 2017-04-04 Abbvie Deutschland Gmbh & Co Kg Powdered protein compositions and methods of making same
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
WO2017136433A1 (en) * 2016-02-03 2017-08-10 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US9878102B2 (en) 2011-01-24 2018-01-30 Abbvie Biotechnology Ltd. Automatic injection devices having overmolded gripping surfaces
RU2665966C2 (ru) * 2016-12-30 2018-09-05 Закрытое Акционерное Общество "Биокад" Водная фармацевтическая композиция рекомбинантного моноклонального антитела к ФНОα
US10125191B2 (en) 2005-05-11 2018-11-13 Alexion Pharmaceutiacls, Inc. Compositions comprising an anti-C5 antibody
WO2019057631A1 (en) 2017-09-20 2019-03-28 Alvotech Hf PHARMACEUTICAL FORMULATIONS FOR ADALIMUMAB
EP2676677B1 (en) * 2011-02-17 2019-05-22 Kyowa Hakko Kirin Co., Ltd. Highly concentrated anti-cd40 antibody pharmaceutical preparation
US10322176B2 (en) 2002-03-01 2019-06-18 Immunomedics, Inc. Subcutaneous administration of anti-CD74 antibody for systemic lupus erythematosus
US10493152B2 (en) 2014-05-23 2019-12-03 Fresenius Kabi Deutschland Gmbh Adalimumab formulations
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
US10799597B2 (en) 2017-04-03 2020-10-13 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy
US10918698B2 (en) * 2014-03-29 2021-02-16 Intas Pharmaceuticals Ltd. Lyophilized pharmaceutical composition of Fc-peptide fusion protein
US11071782B2 (en) 2016-04-20 2021-07-27 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11180559B2 (en) 2005-03-03 2021-11-23 Immunomedics, Inc. Subcutaneous anti-HLA-DR monoclonal antibody for treatment of hematologic malignancies
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11333642B2 (en) 2016-10-25 2022-05-17 Regeneran Pharmaceuticals, Inc. Methods and systems for chromatography data analysis
US11369896B2 (en) 2016-08-16 2022-06-28 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11633476B2 (en) 2017-05-02 2023-04-25 Merck Sharp & Dohme Llc Stable formulations of programmed death receptor 1 (PD-1) antibodies and methods of use thereof
US11845798B2 (en) 2017-05-02 2023-12-19 Merck Sharp & Dohme Llc Formulations of anti-LAG3 antibodies and co-formulations of anti-LAG3 antibodies and anti-PD-1 antibodies
US11884698B2 (en) 2018-07-02 2024-01-30 Regeneron Pharmaceuticals, Inc. Systems and methods for preparing a polypeptide from a mixture

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG10201609982PA (en) 2012-03-07 2017-01-27 Cadila Healthcare Ltd Pharmaceutical formulations of tnf-alpha anitbodies
JP2015519382A (ja) * 2012-06-12 2015-07-09 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 治療用抗体のための医薬処方物
US8883979B2 (en) * 2012-08-31 2014-11-11 Bayer Healthcare Llc Anti-prolactin receptor antibody formulations
CN102988984B (zh) * 2012-12-21 2015-05-20 嘉和生物药业有限公司 增强稳定性的抗TNF-α人单克隆抗体的含水药物制剂
EP2968538A1 (en) 2013-03-15 2016-01-20 Bayer HealthCare LLC Anti-prolactin receptor antibody formulations
CN103446583B (zh) * 2013-03-21 2015-11-18 百奥泰生物科技(广州)有限公司 一种治疗TNF-α相关疾病的人抗体制剂
RU2662558C2 (ru) * 2013-07-19 2018-07-26 Хексаль Аг Способы и композиции, которые обеспечивают модуляцию иммунных ответов, связанных с введением биофармацевтического лекарственного средства
KR102435648B1 (ko) * 2013-09-11 2022-08-25 이글 바이오로직스 인코퍼레이티드 점도저하제를 함유하는 액체 단백질 제형
ES2893861T3 (es) * 2013-10-24 2022-02-10 Astrazeneca Ab Formulaciones de anticuerpo estables, acuosas
CN104666242B (zh) * 2013-11-26 2018-01-02 信达生物制药(苏州)有限公司 一种稳定的抗TNF‑α抗体制剂及其用途
AU2014354384B2 (en) * 2013-11-29 2018-11-08 Ares Trading S.A. A liquid formulation of a fusion protein comprising TNFR and Fc region
AU2015267052A1 (en) * 2014-05-27 2016-12-15 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
KR20170018810A (ko) * 2014-06-10 2017-02-20 메이지 세이카 파루마 가부시키가이샤 안정한 아달리무맙 수성 제제
FI3479819T3 (fi) * 2016-06-30 2024-04-17 Celltrion Inc Stabiili nestemäinen farmaseuttinen valmiste
US11608357B2 (en) 2018-08-28 2023-03-21 Arecor Limited Stabilized antibody protein solutions
EP3372241A1 (en) * 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
EP3372242A1 (en) * 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
CN110832075A (zh) 2017-03-22 2020-02-21 诺华股份有限公司 用于免疫肿瘤学的组合物和方法
CN107485713B (zh) * 2017-07-31 2018-08-28 百奥泰生物科技(广州)有限公司 针对TNF-α的抗体组合物及其应用
KR20190024572A (ko) * 2017-08-30 2019-03-08 (주)셀트리온 TNFα 관련 질환을 치료하기 위한 피하 투여 요법
KR102545825B1 (ko) 2017-09-30 2023-06-22 바이오셀즈(베이징) 바이오테크 코., 엘티디. 흥분성 신경 독성 관련 손상 치료를 위한 펩타이드 조성물
EA201900360A1 (ru) * 2017-12-29 2019-12-30 Закрытое Акционерное Общество "Биокад" ВОДНАЯ ФАРМАЦЕВТИЧЕСКАЯ КОМПОЗИЦИЯ РЕКОМБИНАНТНОГО МОНОКЛОНАЛЬНОГО АНТИТЕЛА К ФНОα
MX2020009935A (es) * 2018-03-23 2021-01-08 Abbvie Deutschland Formulaciones acuosas y estables de anticuerpos anti-tau.
EP4153234A1 (en) * 2020-05-21 2023-03-29 Shilpa Biologicals Private Limited Pharmaceutical compositions comprising adalimumab

Citations (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5237054A (en) * 1987-02-20 1993-08-17 Akzo Pharma Stabilized aqueous composition containing antibodies
US5654403A (en) * 1991-10-28 1997-08-05 Burroughs Wellcome Co. Immunoglobulins stabilized with a chelator of copper ions
US5945098A (en) * 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
US6024938A (en) * 1994-07-07 2000-02-15 Ortho Pharmaceutical Corporation Lyophilized imaging agent formulation comprising a chemotactic peptide
US6090382A (en) * 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US6171586B1 (en) * 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6252055B1 (en) * 1996-05-24 2001-06-26 Glaxo Wellcome Inc. Concentrated antibody preparation
US6258562B1 (en) * 1996-02-09 2001-07-10 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US20030012786A1 (en) * 2001-05-25 2003-01-16 Teoh Leah S. Use of anti-TNF antibodies as drugs in treating septic disorders of anemic patients
US20030045454A1 (en) * 2001-06-21 2003-03-06 Franklin Okumu Sustained release formulation
US20030049725A1 (en) * 2000-08-07 2003-03-13 George Heavner Anti-TNF antibodies, compositions, methods and uses
US20030113316A1 (en) * 2001-07-25 2003-06-19 Kaisheva Elizabet A. Stable lyophilized pharmaceutical formulation of IgG antibodies
US20030138417A1 (en) * 2001-11-08 2003-07-24 Kaisheva Elizabet A. Stable liquid pharmaceutical formulation of IgG antibodies
US20030143603A1 (en) * 2001-11-30 2003-07-31 Jill Giles-Komar Anti-TNF antibodies, compositions, methods and uses
US20030161828A1 (en) * 2002-02-19 2003-08-28 Abbott Gmbh & Co. Kg Use of TNF antagonists as drugs for the treatment of patients with an inflammatory reaction and without suffering from total organ failure
US20040009172A1 (en) * 2002-04-26 2004-01-15 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20040028667A1 (en) * 2000-08-29 2004-02-12 Daniel Norman Stabilisation of immunoglobulins at a low ph
US20040033228A1 (en) * 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
US20040038878A1 (en) * 2000-08-04 2004-02-26 Masahiko Tanikawa Injectable protein formulations
US6737405B2 (en) * 2000-05-08 2004-05-18 Aventis Behring Gmbh Stabilized protein preparation and process for its preparation
US20040126372A1 (en) * 2002-07-19 2004-07-01 Abbott Biotechnology Ltd. Treatment of TNFalpha related disorders
US20040156835A1 (en) * 2001-05-30 2004-08-12 Taiji Imoto Protein preparation
US20040166111A1 (en) * 2002-10-24 2004-08-26 Zehra Kaymakcalan Low dose methods for treating disorders in which TNFalpha activity is detrimental
US20050053598A1 (en) * 2003-02-10 2005-03-10 Burke David J. Immunoglobulin formulation and method of preparation thereof
US6875432B2 (en) * 2000-10-12 2005-04-05 Genentech, Inc. Reduced-viscosity concentrated protein formulations
US20050118163A1 (en) * 2002-02-14 2005-06-02 Hidefumi Mizushima Antibody-containing solution pharmaceuticals
US20050119172A1 (en) * 2002-02-05 2005-06-02 Werner Merkle Stabilization of protein preparations
US20050118167A1 (en) * 1999-04-28 2005-06-02 Yamanouchi Pharmaceutical Co., Ltd. Parenteral pharmaceutical composition containing humanized monoclonal antibody fragment and stabilizing method thereof
US20050175611A1 (en) * 2003-11-26 2005-08-11 Hanns-Christian Mahler Pharmaceutical preparation comprising an antibody against the EGF receptor
US20060009365A1 (en) * 2004-07-08 2006-01-12 Erhan Sevim Z Poly(hydroxy thioether) vegetable oil derivatives useful as lubricant additives
US20060082741A1 (en) * 2004-10-18 2006-04-20 Asml Netherlands B.V. Lithographic apparatus and device manufacturing method
US20060088523A1 (en) * 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulations
US20060115472A1 (en) * 2004-08-13 2006-06-01 Wyeth Stabilizing formulations
US7070775B2 (en) * 1991-03-18 2006-07-04 New York University Recombinant A2-specific TNFα specific antibodies
US20060159653A1 (en) * 2003-02-28 2006-07-20 Chugai Seiyaku Kabushiki Kaisha Stabilized preparation containing protein
US20060182740A1 (en) * 2002-06-21 2006-08-17 Biogen Idec, Inc. Buffered formulations for concentrating antibodies and methods of use thereof
US20070020255A1 (en) * 2003-10-01 2007-01-25 Kyowa Hakko Kogyo Co., Ltd. Method of stabilizing antibody and stabilized solution-type antibody preparation
US20070031402A1 (en) * 2005-08-03 2007-02-08 Immunogen Inc. Immunoconjugate formulations
US20070036779A1 (en) * 2003-04-09 2007-02-15 Annie Bardat Stabilising formulation for immunoglobulin g compositions in liquid form and in lyophilised form
US20070041905A1 (en) * 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
US20070053900A1 (en) * 2003-04-04 2007-03-08 Genentech, Inc. High concentration antibody and protein formulations
US20070053871A1 (en) * 2005-08-05 2007-03-08 Amgen Inc. Pharmaceutical formulations
US20070053906A1 (en) * 2003-02-28 2007-03-08 Ares Trading S.A. Liquid formulations of tumor necrosis factor-binding proteins
US20070071747A1 (en) * 2005-05-16 2007-03-29 Hoffman Rebecca S Use of TNFalpha inhibitor for treatment of erosive polyarthritis
US20070184050A1 (en) * 2003-12-25 2007-08-09 Kirin Beer Kabushiki Kaisha Stable water-based medicinal preparation containing antibody
US20070190047A1 (en) * 2005-07-29 2007-08-16 Amgen, Inc. Formulations that inhibit protein aggregation
US20070197439A1 (en) * 2006-02-07 2007-08-23 Gaozhong Zhu Stabilized compositions of proteins having a free thiol moiety
US20080003220A1 (en) * 2006-04-21 2008-01-03 Amgen, Inc. Buffering agents for biopharmaceutical formulations
US20080071063A1 (en) * 2006-02-03 2008-03-20 Medimmune, Inc. Protein Formulations
US20080112953A1 (en) * 2006-10-06 2008-05-15 Amgen Inc. Stable formulations
US20080118496A1 (en) * 2006-04-10 2008-05-22 Medich John R Uses and compositions for treatment of juvenile rheumatoid arthritis
US20080124326A1 (en) * 2006-10-20 2008-05-29 Amgen Inc. Stable polypeptide formulations
US20080131374A1 (en) * 2006-04-19 2008-06-05 Medich John R Uses and compositions for treatment of rheumatoid arthritis
US20080139792A1 (en) * 2006-12-06 2008-06-12 Wyeth High Protein Concentration Formulations Containing Mannitol
US20080145383A1 (en) * 2004-03-12 2008-06-19 Intercell Ag Method for Solubilizing Peptide Mixtures
US20080166348A1 (en) * 2006-04-10 2008-07-10 Hartmut Kupper Uses and compositions for treatment of rheumatoid arthritis
US20090017472A1 (en) * 2007-05-31 2009-01-15 Bruno Stuhlmuller BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNFalpha INHIBITORS IN AUTOIMMUNE DISORDERS
US20090029794A1 (en) * 2007-07-23 2009-01-29 Yung-Hsiung Chen Golf Club Head that Reduces a Contact Resistance with the Ground
US20090054331A1 (en) * 2007-06-01 2009-02-26 Acologix, Inc. High temperature stable peptide formulation
US20090110679A1 (en) * 2007-07-13 2009-04-30 Luk-Chiu Li Methods and compositions for pulmonary administration of a TNFa inhibitor
US20090123378A1 (en) * 2006-04-10 2009-05-14 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20090148406A1 (en) * 2005-07-02 2009-06-11 Arecor Limited Stable Aqueous Systems Comprising Proteins
US20090148513A1 (en) * 2007-08-08 2009-06-11 Wolfgang Fraunhofer Compositions and methods for crystallizing antibodies
US20100003243A1 (en) * 2007-06-01 2010-01-07 Okun Martin M Uses and Compositions for treatment of Psoriasis and Crohn's Disease
US20100021451A1 (en) * 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20100028383A1 (en) * 2005-03-08 2010-02-04 Intervet International B.V. Chemically defined stabiliser
US20100028372A1 (en) * 2007-01-11 2010-02-04 Jan Jezek Stabilization of aqueous compositions of proteins with displacement buffers
US20100034823A1 (en) * 2006-10-27 2010-02-11 Borhani David W Crystalline anti-hTNFalpha antibodies
US20100040630A1 (en) * 2008-03-24 2010-02-18 Aake Elden Methods and compositions for treating bone loss
US20100113744A1 (en) * 2007-04-26 2010-05-06 Bayer Healthcare Llc Stabilization Of Liquid Solutions Of Recombinant Protein For Frozen Storage
US20100129379A1 (en) * 2006-09-25 2010-05-27 John Carpenter Stabilized antibody formulations and uses thereof
US20100160894A1 (en) * 2006-06-30 2010-06-24 Julian Joseph F Automatic injection device
US7758860B2 (en) * 2005-12-21 2010-07-20 Wyeth Llc Protein formulations with reduced viscosity and uses thereof
US7863426B2 (en) * 2006-04-05 2011-01-04 Abbott Biotechnology Ltd. Antibody purification
US20110014676A1 (en) * 2007-06-29 2011-01-20 Battelle Memorial Institute Protein stabilization
US20110054414A1 (en) * 2009-04-29 2011-03-03 Abbott Biotechnology Ltd. Automatic Injection Device
US7919264B2 (en) * 2005-11-01 2011-04-05 Abbott Biotechnology Ltd. Methods and compositions for determining the efficacy of a treatment for ankylosing spondylitis using biomarkers
US20110171227A1 (en) * 2006-04-10 2011-07-14 Okun Martin M Methods and compositions for treatment of skin disorders
US20120014956A1 (en) * 2010-02-02 2012-01-19 Hartmut Kupper Methods and compositions for predicting responsiveness to treatment with tnf-alpha inhibitor

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9803710L (sv) * 1998-09-25 2000-03-26 A & Science Invest Ab Användning av vissa substanser för behandling av nervrotsskador
CA2604840A1 (en) * 2005-04-18 2006-10-26 Xtl Biopharmaceuticals Ltd. Stabilized anti-hepatitis b (hbv) antibody formulations

Patent Citations (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5237054A (en) * 1987-02-20 1993-08-17 Akzo Pharma Stabilized aqueous composition containing antibodies
US5945098A (en) * 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
US7070775B2 (en) * 1991-03-18 2006-07-04 New York University Recombinant A2-specific TNFα specific antibodies
US5654403A (en) * 1991-10-28 1997-08-05 Burroughs Wellcome Co. Immunoglobulins stabilized with a chelator of copper ions
US6024938A (en) * 1994-07-07 2000-02-15 Ortho Pharmaceutical Corporation Lyophilized imaging agent formulation comprising a chemotactic peptide
US6509015B1 (en) * 1996-02-09 2003-01-21 Basf Aktiengesellschaft Human antibodies that bind human TNFa
US6258562B1 (en) * 1996-02-09 2001-07-10 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US20100040604A1 (en) * 1996-02-09 2010-02-18 Salfeld Jochen G HUMAN ANTIBODIES THAT BIND HUMAN TNFalpha
US6090382A (en) * 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
US7223394B2 (en) * 1996-02-09 2007-05-29 Abbott Biotechnology Ltd Human antibodies that bind human TNFα
US20100016557A1 (en) * 1996-02-09 2010-01-21 Abbott Biotechnology Ltd. HUMAN ANTIBODIES THAT BIND HUMAN TNFalpha
US7541031B2 (en) * 1996-02-09 2009-06-02 Abbott Biotechnology Ltd. Methods for treating rheumatoid arthritis using human antibodies that bind human TNFα
US20090155205A1 (en) * 1996-02-09 2009-06-18 Salfeld Jochen G HUMAN ANTIBODIES THAT BIND HUMAN TNFa
US6252055B1 (en) * 1996-05-24 2001-06-26 Glaxo Wellcome Inc. Concentrated antibody preparation
US6171586B1 (en) * 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US20050118167A1 (en) * 1999-04-28 2005-06-02 Yamanouchi Pharmaceutical Co., Ltd. Parenteral pharmaceutical composition containing humanized monoclonal antibody fragment and stabilizing method thereof
US6737405B2 (en) * 2000-05-08 2004-05-18 Aventis Behring Gmbh Stabilized protein preparation and process for its preparation
US20040038878A1 (en) * 2000-08-04 2004-02-26 Masahiko Tanikawa Injectable protein formulations
US7250165B2 (en) * 2000-08-07 2007-07-31 Centocor, Inc. Anti-TNF antibodies, compositions, methods and uses
US20030049725A1 (en) * 2000-08-07 2003-03-13 George Heavner Anti-TNF antibodies, compositions, methods and uses
US7220409B2 (en) * 2000-08-29 2007-05-22 Probi Ab Stabilization of immunoglobulins at low pH
US20040028667A1 (en) * 2000-08-29 2004-02-12 Daniel Norman Stabilisation of immunoglobulins at a low ph
US6875432B2 (en) * 2000-10-12 2005-04-05 Genentech, Inc. Reduced-viscosity concentrated protein formulations
US20030012786A1 (en) * 2001-05-25 2003-01-16 Teoh Leah S. Use of anti-TNF antibodies as drugs in treating septic disorders of anemic patients
US20040156835A1 (en) * 2001-05-30 2004-08-12 Taiji Imoto Protein preparation
US20030045454A1 (en) * 2001-06-21 2003-03-06 Franklin Okumu Sustained release formulation
US7318931B2 (en) * 2001-06-21 2008-01-15 Genentech, Inc. Sustained release formulation
US20030113316A1 (en) * 2001-07-25 2003-06-19 Kaisheva Elizabet A. Stable lyophilized pharmaceutical formulation of IgG antibodies
US20030138417A1 (en) * 2001-11-08 2003-07-24 Kaisheva Elizabet A. Stable liquid pharmaceutical formulation of IgG antibodies
US20030143603A1 (en) * 2001-11-30 2003-07-31 Jill Giles-Komar Anti-TNF antibodies, compositions, methods and uses
US20050119172A1 (en) * 2002-02-05 2005-06-02 Werner Merkle Stabilization of protein preparations
US20050118163A1 (en) * 2002-02-14 2005-06-02 Hidefumi Mizushima Antibody-containing solution pharmaceuticals
US20030161828A1 (en) * 2002-02-19 2003-08-28 Abbott Gmbh & Co. Kg Use of TNF antagonists as drugs for the treatment of patients with an inflammatory reaction and without suffering from total organ failure
US20040009172A1 (en) * 2002-04-26 2004-01-15 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20060182740A1 (en) * 2002-06-21 2006-08-17 Biogen Idec, Inc. Buffered formulations for concentrating antibodies and methods of use thereof
US20040131614A1 (en) * 2002-07-19 2004-07-08 Abbott Biotechnology Ltd. Treatment of pulmonary disorders using TNFalpha inhibitor
US20040136989A1 (en) * 2002-07-19 2004-07-15 Abbott Laboratories S.A. Treatment of vasculitides using TNFalpha inhibitors
US20070202104A1 (en) * 2002-07-19 2007-08-30 Abbott Laboratories S.A. Treatment of spondyloarthropathies using TNFalpha inhibitors
US20040126373A1 (en) * 2002-07-19 2004-07-01 Abbott Biotechnology Ltd. Treatment of coronary disorders using TNFalpha inhibitors
US20040126372A1 (en) * 2002-07-19 2004-07-01 Abbott Biotechnology Ltd. Treatment of TNFalpha related disorders
US20040136991A1 (en) * 2002-07-19 2004-07-15 Abbott Biotechnology Ltd. Treatment of anemia using TNFalpha inhibitors
US20040136990A1 (en) * 2002-07-19 2004-07-15 Abbott Biotechnology Ltd. Treatment of pain using TNFalpha inhibitors
US20040151722A1 (en) * 2002-07-19 2004-08-05 Abbott Biotechnology Ltd. Treatment of metabolic disorders using TNFalpha inhibitors
US20040033228A1 (en) * 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
US20060153846A1 (en) * 2002-08-16 2006-07-13 Hans-Juergen Krause Formulation of human antibodies for treating tnf-alpha associated disorders
US8216583B2 (en) * 2002-08-16 2012-07-10 Abbott Biotechnology, Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US20040166111A1 (en) * 2002-10-24 2004-08-26 Zehra Kaymakcalan Low dose methods for treating disorders in which TNFalpha activity is detrimental
US20050053598A1 (en) * 2003-02-10 2005-03-10 Burke David J. Immunoglobulin formulation and method of preparation thereof
US20070053906A1 (en) * 2003-02-28 2007-03-08 Ares Trading S.A. Liquid formulations of tumor necrosis factor-binding proteins
US20060159653A1 (en) * 2003-02-28 2006-07-20 Chugai Seiyaku Kabushiki Kaisha Stabilized preparation containing protein
US20070053900A1 (en) * 2003-04-04 2007-03-08 Genentech, Inc. High concentration antibody and protein formulations
US20070036779A1 (en) * 2003-04-09 2007-02-15 Annie Bardat Stabilising formulation for immunoglobulin g compositions in liquid form and in lyophilised form
US20070020255A1 (en) * 2003-10-01 2007-01-25 Kyowa Hakko Kogyo Co., Ltd. Method of stabilizing antibody and stabilized solution-type antibody preparation
US20050175611A1 (en) * 2003-11-26 2005-08-11 Hanns-Christian Mahler Pharmaceutical preparation comprising an antibody against the EGF receptor
US20070184050A1 (en) * 2003-12-25 2007-08-09 Kirin Beer Kabushiki Kaisha Stable water-based medicinal preparation containing antibody
US20080145383A1 (en) * 2004-03-12 2008-06-19 Intercell Ag Method for Solubilizing Peptide Mixtures
US20060009365A1 (en) * 2004-07-08 2006-01-12 Erhan Sevim Z Poly(hydroxy thioether) vegetable oil derivatives useful as lubricant additives
US20060115472A1 (en) * 2004-08-13 2006-06-01 Wyeth Stabilizing formulations
US20060082741A1 (en) * 2004-10-18 2006-04-20 Asml Netherlands B.V. Lithographic apparatus and device manufacturing method
US20060088523A1 (en) * 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulations
US20100028383A1 (en) * 2005-03-08 2010-02-04 Intervet International B.V. Chemically defined stabiliser
US20070071747A1 (en) * 2005-05-16 2007-03-29 Hoffman Rebecca S Use of TNFalpha inhibitor for treatment of erosive polyarthritis
US20090148406A1 (en) * 2005-07-02 2009-06-11 Arecor Limited Stable Aqueous Systems Comprising Proteins
US20070190047A1 (en) * 2005-07-29 2007-08-16 Amgen, Inc. Formulations that inhibit protein aggregation
US20070031402A1 (en) * 2005-08-03 2007-02-08 Immunogen Inc. Immunoconjugate formulations
US20070053871A1 (en) * 2005-08-05 2007-03-08 Amgen Inc. Pharmaceutical formulations
US20070081996A1 (en) * 2005-08-19 2007-04-12 Hoffman Rebecca S Method of treating depression using a TNFalpha antibody
US20070041905A1 (en) * 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
US7919264B2 (en) * 2005-11-01 2011-04-05 Abbott Biotechnology Ltd. Methods and compositions for determining the efficacy of a treatment for ankylosing spondylitis using biomarkers
US7758860B2 (en) * 2005-12-21 2010-07-20 Wyeth Llc Protein formulations with reduced viscosity and uses thereof
US20080071063A1 (en) * 2006-02-03 2008-03-20 Medimmune, Inc. Protein Formulations
US20070197439A1 (en) * 2006-02-07 2007-08-23 Gaozhong Zhu Stabilized compositions of proteins having a free thiol moiety
US20110002935A1 (en) * 2006-04-05 2011-01-06 Min Wan Antibody purification
US7863426B2 (en) * 2006-04-05 2011-01-04 Abbott Biotechnology Ltd. Antibody purification
US20090123378A1 (en) * 2006-04-10 2009-05-14 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20080166348A1 (en) * 2006-04-10 2008-07-10 Hartmut Kupper Uses and compositions for treatment of rheumatoid arthritis
US20110171227A1 (en) * 2006-04-10 2011-07-14 Okun Martin M Methods and compositions for treatment of skin disorders
US20080118496A1 (en) * 2006-04-10 2008-05-22 Medich John R Uses and compositions for treatment of juvenile rheumatoid arthritis
US20080131374A1 (en) * 2006-04-19 2008-06-05 Medich John R Uses and compositions for treatment of rheumatoid arthritis
US20080003220A1 (en) * 2006-04-21 2008-01-03 Amgen, Inc. Buffering agents for biopharmaceutical formulations
US20100021451A1 (en) * 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20100160894A1 (en) * 2006-06-30 2010-06-24 Julian Joseph F Automatic injection device
US20100129379A1 (en) * 2006-09-25 2010-05-27 John Carpenter Stabilized antibody formulations and uses thereof
US20080112953A1 (en) * 2006-10-06 2008-05-15 Amgen Inc. Stable formulations
US20080124326A1 (en) * 2006-10-20 2008-05-29 Amgen Inc. Stable polypeptide formulations
US20100034823A1 (en) * 2006-10-27 2010-02-11 Borhani David W Crystalline anti-hTNFalpha antibodies
US20080139792A1 (en) * 2006-12-06 2008-06-12 Wyeth High Protein Concentration Formulations Containing Mannitol
US20100028372A1 (en) * 2007-01-11 2010-02-04 Jan Jezek Stabilization of aqueous compositions of proteins with displacement buffers
US20100113744A1 (en) * 2007-04-26 2010-05-06 Bayer Healthcare Llc Stabilization Of Liquid Solutions Of Recombinant Protein For Frozen Storage
US20120039900A1 (en) * 2007-05-31 2012-02-16 Abbott Laboratories Biomarkers predictive of the responsiveness to tnfalpha inhibitors in autoimmune disorders
US20090017472A1 (en) * 2007-05-31 2009-01-15 Bruno Stuhlmuller BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNFalpha INHIBITORS IN AUTOIMMUNE DISORDERS
US20100003243A1 (en) * 2007-06-01 2010-01-07 Okun Martin M Uses and Compositions for treatment of Psoriasis and Crohn's Disease
US20090054331A1 (en) * 2007-06-01 2009-02-26 Acologix, Inc. High temperature stable peptide formulation
US20110014676A1 (en) * 2007-06-29 2011-01-20 Battelle Memorial Institute Protein stabilization
US20090110679A1 (en) * 2007-07-13 2009-04-30 Luk-Chiu Li Methods and compositions for pulmonary administration of a TNFa inhibitor
US20090029794A1 (en) * 2007-07-23 2009-01-29 Yung-Hsiung Chen Golf Club Head that Reduces a Contact Resistance with the Ground
US20090148513A1 (en) * 2007-08-08 2009-06-11 Wolfgang Fraunhofer Compositions and methods for crystallizing antibodies
US20100040630A1 (en) * 2008-03-24 2010-02-18 Aake Elden Methods and compositions for treating bone loss
US20110054414A1 (en) * 2009-04-29 2011-03-03 Abbott Biotechnology Ltd. Automatic Injection Device
US20120014956A1 (en) * 2010-02-02 2012-01-19 Hartmut Kupper Methods and compositions for predicting responsiveness to treatment with tnf-alpha inhibitor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Sarciaux et al. (J. of Pharmaceutical Sciences, Vol. 88, No. 12, December 1999) *
Weinblatt et al. (Arthritis & Rheumatism, Vol. 48, No.1, pp. 35-45, January 2003) *

Cited By (285)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100016557A1 (en) * 1996-02-09 2010-01-21 Abbott Biotechnology Ltd. HUMAN ANTIBODIES THAT BIND HUMAN TNFalpha
US8206714B2 (en) 1996-02-09 2012-06-26 Abbott Biotechnology Ltd. Methods for treating rheumatoid arthritis using human antibodies that bind human TNFa
US8753633B2 (en) 1996-02-09 2014-06-17 Abbvie Biotechnology Ltd. Human antibodies that bind human TNFα
US8372400B2 (en) 1996-02-09 2013-02-12 Abbott Biotechnology Ltd. Methods of treating disorders using human antibodies that bind human TNFα
US8197813B2 (en) 1996-02-09 2012-06-12 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα
US8372401B2 (en) 1996-02-09 2013-02-12 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα
US20090155205A1 (en) * 1996-02-09 2009-06-18 Salfeld Jochen G HUMAN ANTIBODIES THAT BIND HUMAN TNFa
US8414894B2 (en) 1996-02-09 2013-04-09 Abbott Biotechnology Ltd. Human antibodies that bind human TNFα and methods of using same
US9340612B2 (en) 2001-03-19 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US20030235585A1 (en) * 2001-06-08 2003-12-25 Fischkoff Steven A. Methods of administering anti-TNFalpha antibodies
US8992926B2 (en) 2001-06-08 2015-03-31 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8974790B2 (en) 2001-06-08 2015-03-10 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US9017680B2 (en) 2001-06-08 2015-04-28 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8911737B2 (en) 2001-06-08 2014-12-16 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US9073987B2 (en) 2001-06-08 2015-07-07 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US8889135B2 (en) 2001-06-08 2014-11-18 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US9546212B2 (en) 2001-06-08 2017-01-17 Abbvie Biotechnology Ltd. Methods of administering anti-TNFα antibodies
US10322176B2 (en) 2002-03-01 2019-06-18 Immunomedics, Inc. Subcutaneous administration of anti-CD74 antibody for systemic lupus erythematosus
US9085620B1 (en) 2002-07-19 2015-07-21 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriatic arthritis
US8906373B2 (en) 2002-07-19 2014-12-09 Abbvie Biotechnology Ltd. Use of TNF-alpha inhibitor for treatment of psoriasis
US9090689B1 (en) 2002-07-19 2015-07-28 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriasis
US20040126372A1 (en) * 2002-07-19 2004-07-01 Abbott Biotechnology Ltd. Treatment of TNFalpha related disorders
US9750808B2 (en) 2002-08-16 2017-09-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US9738714B2 (en) 2002-08-16 2017-08-22 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8216583B2 (en) 2002-08-16 2012-07-10 Abbott Biotechnology, Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9295725B2 (en) 2002-08-16 2016-03-29 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9289497B2 (en) 2002-08-16 2016-03-22 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9272041B2 (en) 2002-08-16 2016-03-01 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9272042B2 (en) 2002-08-16 2016-03-01 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9327032B2 (en) 2002-08-16 2016-05-03 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8802101B2 (en) 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9220781B2 (en) 2002-08-16 2015-12-29 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8802100B2 (en) 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US20060153846A1 (en) * 2002-08-16 2006-07-13 Hans-Juergen Krause Formulation of human antibodies for treating tnf-alpha associated disorders
US9732152B2 (en) 2002-08-16 2017-08-15 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8795670B2 (en) 2002-08-16 2014-08-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US9114166B2 (en) 2002-08-16 2015-08-25 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9302011B2 (en) 2002-08-16 2016-04-05 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-α associated disorders
US8802102B2 (en) 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9950066B2 (en) 2002-08-16 2018-04-24 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8940305B2 (en) 2002-08-16 2015-01-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8932591B2 (en) 2002-08-16 2015-01-13 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8916158B2 (en) 2002-08-16 2014-12-23 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8916157B2 (en) 2002-08-16 2014-12-23 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8911741B2 (en) 2002-08-16 2014-12-16 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US20140056888A1 (en) * 2002-09-06 2014-02-27 Alexion Pharmaceuticals, Inc. High concentration antibody formulations
US9415102B2 (en) 2002-09-06 2016-08-16 Alexion Pharmaceuticals, Inc. High concentration formulations of anti-C5 antibodies
US9352035B2 (en) * 2002-09-06 2016-05-31 Alexion Pharmaceuticals, Inc. High concentration antibody formulations
US8961973B2 (en) 2004-04-09 2015-02-24 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US9061005B2 (en) 2004-04-09 2015-06-23 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating idiopathic inflammatory bowel disease
US8961974B2 (en) 2004-04-09 2015-02-24 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US9499615B2 (en) 2004-04-09 2016-11-22 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating idiopathic inflammatory bowel disease
US9512216B2 (en) 2004-04-09 2016-12-06 Abbvie Biotechnology Ltd. Use of TNFα inhibitor
US9187559B2 (en) 2004-04-09 2015-11-17 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating idiopathic inflammatory bowel disease
US20090304682A1 (en) * 2004-04-09 2009-12-10 Hoffman Rebecca S Multiple-variable dose regimen for treating TNFa-related disorders
US8986693B1 (en) 2004-04-09 2015-03-24 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriasis
US8889136B2 (en) 2004-04-09 2014-11-18 Abbvie Biotechnology Ltd. Multiple-variable dose regimen for treating TNFα-related disorders
US8162887B2 (en) 2004-06-23 2012-04-24 Abbott Biotechnology Ltd. Automatic injection devices
US8668670B2 (en) 2004-06-23 2014-03-11 Abbvie Biotechnology Ltd Automatic injection devices
US9017287B2 (en) 2004-06-23 2015-04-28 Abbvie Biotechnology Ltd Automatic injection devices
US20060083741A1 (en) * 2004-10-08 2006-04-20 Hoffman Rebecca S Treatment of respiratory syncytial virus (RSV) infection
US11180559B2 (en) 2005-03-03 2021-11-23 Immunomedics, Inc. Subcutaneous anti-HLA-DR monoclonal antibody for treatment of hematologic malignancies
US10125191B2 (en) 2005-05-11 2018-11-13 Alexion Pharmaceutiacls, Inc. Compositions comprising an anti-C5 antibody
US10479828B2 (en) 2005-05-11 2019-11-19 Alexion Pharmaceuticals, Inc. Compositions comprising an anti-C5 antibody
US10927166B2 (en) 2005-05-11 2021-02-23 Alexion Pharmaceuticals, Inc. Compositions comprising an anti-C5 antibody
US9067992B2 (en) 2005-05-16 2015-06-30 Abbvie Biotechnology Ltd. Use of TNFα inhibitor for treatment of psoriatic arthritis
US8808700B1 (en) 2005-05-16 2014-08-19 Abbvie Biotechnology Ltd. Use of TNF alpha inhibitor for treatment of erosive polyarthritis
US8715664B2 (en) 2005-05-16 2014-05-06 Abbvie Biotechnology Ltd. Use of human TNFα antibodies for treatment of erosive polyarthritis
US9086418B2 (en) 2005-11-01 2015-07-21 Abbvie Biotechnology Ltd. Methods and compositions for diagnosing ankylosing spondylitis using biomarkers
US8916153B2 (en) 2006-04-05 2014-12-23 Abbvie Biotechnology Ltd. Purified antibody composition
US9102723B2 (en) 2006-04-05 2015-08-11 Abbvie Biotechnology Ltd Purified antibody composition
US8231876B2 (en) 2006-04-05 2012-07-31 Abbott Biotechnology Ltd. Purified antibody composition
US9328165B2 (en) 2006-04-05 2016-05-03 Abbvie Biotechnology Ltd. Purified antibody composition
US9273132B2 (en) 2006-04-05 2016-03-01 Abbvie Biotechnology Ltd Purified antibody composition
US9096666B2 (en) 2006-04-05 2015-08-04 Abbvie Biotechnology Ltd Purified antibody composition
US11083792B2 (en) 2006-04-05 2021-08-10 Abbvie Biotechnology Ltd Purified antibody composition
US9913902B2 (en) 2006-04-05 2018-03-13 Abbvie Biotechnology Ltd. Purified antibody composition
US8883156B2 (en) 2006-04-05 2014-11-11 Abbvie Biotechnology Ltd. Purified antibody composition
US20110002935A1 (en) * 2006-04-05 2011-01-06 Min Wan Antibody purification
US8895009B2 (en) 2006-04-05 2014-11-25 Abbvie Biotechnology Ltd. Purified antibody composition
US8906372B2 (en) 2006-04-05 2014-12-09 Abbvie Biotechnology Ltd. Purified antibody composition
US9279015B2 (en) 2006-04-10 2016-03-08 Robert L. Wong Methods for treatment of ankylosing spondylitis using TNF alpha antibodies
US20090317399A1 (en) * 2006-04-10 2009-12-24 Pollack Paul F Uses and compositions for treatment of CROHN'S disease
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
US20110171227A1 (en) * 2006-04-10 2011-07-14 Okun Martin M Methods and compositions for treatment of skin disorders
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
US20090280065A1 (en) * 2006-04-10 2009-11-12 Willian Mary K Uses and Compositions for Treatment of Psoriasis
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
US20080311043A1 (en) * 2006-06-08 2008-12-18 Hoffman Rebecca S Uses and compositions for treatment of psoriatic arthritis
US8926975B2 (en) 2006-06-08 2015-01-06 Abbvie Biotechnology Ltd Method of treating ankylosing spondylitis
US20100021451A1 (en) * 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US8679061B2 (en) 2006-06-30 2014-03-25 Abbvie Biotechnology Ltd Automatic injection device
US9486584B2 (en) 2006-06-30 2016-11-08 Abbvie Biotechnology Ltd. Automatic injection device
US8772458B2 (en) 2006-10-27 2014-07-08 Abbvie Biotechnology Ltd Crystalline anti-hTNFalpha antibodies
US8034906B2 (en) 2006-10-27 2011-10-11 Abbott Biotechnology Ltd. Crystalline anti-hTNFalpha antibodies
US8436149B2 (en) 2006-10-27 2013-05-07 Abbvie Biotechnology Ltd Crystalline anti-hTNFalpha antibodies
US20100034823A1 (en) * 2006-10-27 2010-02-11 Borhani David W Crystalline anti-hTNFalpha antibodies
US8404819B2 (en) 2007-03-29 2013-03-26 Abbvie Inc. Crystalline anti-human IL-12 antibodies
US8168760B2 (en) 2007-03-29 2012-05-01 Abbott Laboratories Crystalline anti-human IL-12 antibodies
US20080292642A1 (en) * 2007-03-29 2008-11-27 Borhani David W Crystalline anti-human IL-12 antibodies
US8940873B2 (en) 2007-03-29 2015-01-27 Abbvie Inc. Crystalline anti-human IL-12 antibodies
US9669093B2 (en) 2007-06-11 2017-06-06 Abbvie Biotechnology Ltd Methods for treating juvenile idiopathic arthritis
US9284370B1 (en) 2007-06-11 2016-03-15 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
US20090110679A1 (en) * 2007-07-13 2009-04-30 Luk-Chiu Li Methods and compositions for pulmonary administration of a TNFa inhibitor
US8753839B2 (en) 2007-08-08 2014-06-17 Abbvie Inc. Compositions and methods for crystallizing antibodies
US11191834B2 (en) 2007-11-30 2021-12-07 Abbvie Biotechnology Ltd Protein formulations and methods of making same
US20090291062A1 (en) * 2007-11-30 2009-11-26 Wolfgang Fraunhofer Protein formulations and methods of making same
US9085619B2 (en) 2007-11-30 2015-07-21 Abbvie Biotechnology Ltd. Anti-TNF antibody formulations
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
US8420081B2 (en) 2007-11-30 2013-04-16 Abbvie, Inc. Antibody formulations and methods of making same
US11167030B2 (en) 2007-11-30 2021-11-09 Abbvie Biotechnology Ltd Protein formulations and methods of making same
US20090271164A1 (en) * 2008-01-03 2009-10-29 Peng Joanna Z Predicting long-term efficacy of a compound in the treatment of psoriasis
US9610301B2 (en) 2008-01-15 2017-04-04 Abbvie Deutschland Gmbh & Co Kg Powdered protein compositions and methods of making same
US8636704B2 (en) 2009-04-29 2014-01-28 Abbvie Biotechnology Ltd Automatic injection device
US8758301B2 (en) 2009-12-15 2014-06-24 Abbvie Biotechnology Ltd Firing button for automatic injection device
WO2011097301A2 (en) 2010-02-02 2011-08-11 Abbott Biotechnology Ltd. METHODS AND COMPOSITIONS FOR PREDICTING RESPONSIVENESS TO TREATMENT WITH TNF-α INHIBITOR
US8747854B2 (en) 2010-06-03 2014-06-10 Abbvie Biotechnology Ltd. Methods of treating moderate to severe hidradenitis suppurativa with anti-TNF-alpha antibodies
US9334320B2 (en) 2010-06-03 2016-05-10 Abbvie Biotechnology Ltd. Methods of treating moderate to severe hidradenitis suppurativa with anti-TNFalpha antibody
CN105854016A (zh) * 2010-11-11 2016-08-17 艾伯维生物技术有限公司 改进的高浓度抗TNFα抗体液体制剂
RU2639386C2 (ru) * 2010-11-11 2017-12-21 Эббви Байотекнолоджи Лтд. УЛУЧШЕННЫЕ ВЫСОКОКОНЦЕТРИРОВАННЫЕ ЖИДКИЕ ПРЕПАРАТЫ АНТИТЕЛ ПРОТИВ TNF-альфа
US8821865B2 (en) 2010-11-11 2014-09-02 Abbvie Biotechnology Ltd. High concentration anti-TNFα antibody liquid formulations
EP2637690B1 (en) 2010-11-11 2016-09-21 AbbVie Biotechnology Ltd HIGH CONCENTRATION ANTI-TNFalpha ANTIBODY LIQUID FORMULATIONS
AU2011325974B2 (en) * 2010-11-11 2016-10-27 Abbvie Biotechnology Ltd. Improved high concentration anti-TNFalpha antibody liquid formulations
TWI606840B (zh) * 2010-11-11 2017-12-01 艾伯維生物技術有限責任公司 具有增進高濃度之抗-TNFα抗體之液體調配物
EP2637690A4 (en) * 2010-11-11 2014-04-30 Abbvie Biotechnology Ltd IMPROVED ANTI-TNF-ALPHA-ANTIBODY LIQUID FORMULATIONS WITH HIGH CONCENTRATION
EP2637690A2 (en) * 2010-11-11 2013-09-18 AbbVie Biotechnology Ltd IMPROVED HIGH CONCENTRATION ANTI-TNFalpha ANTIBODY LIQUID FORMULATIONS
CN103458926A (zh) * 2010-11-11 2013-12-18 阿布维生物技术有限公司 改进的高浓度抗TNF α 抗体液体制剂
WO2012065072A3 (en) * 2010-11-11 2012-07-26 Abbott Biotechnology Ltd. IMPROVED HIGH CONCENTRATION ANTI-TNFα ANTIBODY LIQUID FORMULATIONS
US9878102B2 (en) 2011-01-24 2018-01-30 Abbvie Biotechnology Ltd. Automatic injection devices having overmolded gripping surfaces
US11565048B2 (en) 2011-01-24 2023-01-31 Abbvie Biotechnology Ltd. Automatic injection devices having overmolded gripping surfaces
EP2676677B1 (en) * 2011-02-17 2019-05-22 Kyowa Hakko Kirin Co., Ltd. Highly concentrated anti-cd40 antibody pharmaceutical preparation
US9925262B2 (en) 2011-03-08 2018-03-27 Alexion Pharmaceuticals, Inc. Kits comprising formulations of anti-C5 antibodies
US9556263B2 (en) 2011-03-08 2017-01-31 Alexion Pharmaceuticals, Inc. Methods for treating atypical hemolytic uremic syndrome with high concentration formulations of anti-C5 antibodies
US9409980B1 (en) 2011-03-08 2016-08-09 Alexion Pharmaceuticals, Inc. Methods for treating patients with complement-associated disorders with high concentration formulations of anti-C5 antibodies
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9090688B2 (en) 2011-04-27 2015-07-28 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9365645B1 (en) 2011-04-27 2016-06-14 Abbvie, Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9963516B2 (en) 2011-05-02 2018-05-08 Immunomedics, Inc. Stable compositions of high-concentration allotype-selected antibodies for small-volume administration
US8658773B2 (en) * 2011-05-02 2014-02-25 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
US9468689B2 (en) 2011-05-02 2016-10-18 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
US9180205B2 (en) 2011-05-02 2015-11-10 Immunomedics, Inc. Stable compositions of high-concentration allotype-selected antibodies for small-volume administration
US9683050B2 (en) 2011-05-02 2017-06-20 Immunomedics, Inc. Stable compositions of high-concentration allotype-selected antibodies for small-volume administration
EP3009450A1 (en) * 2011-07-19 2016-04-20 Glaxo Group Limited Liquid formulation comprising adalimumab and an acetate buffer
WO2013096835A1 (en) * 2011-12-23 2013-06-27 Abbvie Inc. Stable protein formulations
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9346879B2 (en) 2012-04-20 2016-05-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9359434B2 (en) 2012-04-20 2016-06-07 Abbvie, Inc. Cell culture methods to reduce acidic species
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9290568B2 (en) 2012-09-02 2016-03-22 Abbvie, Inc. Methods to control protein heterogeneity
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
EA029215B1 (ru) * 2012-09-07 2018-02-28 Кохерус Байосайенсис, Инк. Стабильные водные составы адалимумаба
US10772959B2 (en) 2012-09-07 2020-09-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
WO2014039903A2 (en) 2012-09-07 2014-03-13 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
WO2014039903A3 (en) * 2012-09-07 2014-06-26 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10799585B2 (en) 2012-09-07 2020-10-13 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10786566B2 (en) 2012-09-07 2020-09-29 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10780163B2 (en) 2012-09-07 2020-09-22 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10772960B2 (en) 2012-09-07 2020-09-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10722579B2 (en) 2012-09-07 2020-07-28 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716852B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716853B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716854B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10688183B2 (en) 2012-09-07 2020-06-23 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
EP2892550B1 (en) * 2012-09-07 2019-12-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10286072B2 (en) 2012-09-07 2019-05-14 Coherus Biosciences, Inc. Methods of manufacturing stable aqueous formulations of adalimumab
US10286071B2 (en) 2012-09-07 2019-05-14 Coherus Biosciences, Inc. Syringe containing stable aqueous formulations of adalimumab
US10207000B2 (en) 2012-09-07 2019-02-19 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10195275B2 (en) 2012-09-07 2019-02-05 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10159733B2 (en) 2012-09-07 2018-12-25 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10159732B2 (en) 2012-09-07 2018-12-25 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10155039B2 (en) 2012-09-07 2018-12-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9682145B2 (en) 2012-09-07 2017-06-20 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9340611B2 (en) 2012-09-07 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9346880B2 (en) 2012-09-07 2016-05-24 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9861695B2 (en) 2012-09-07 2018-01-09 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9707293B2 (en) 2012-09-07 2017-07-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
CN104768576A (zh) * 2012-09-07 2015-07-08 科荣生生物科学公司 稳定的阿达木单抗水性制剂
US9808525B2 (en) 2012-09-07 2017-11-07 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9724414B2 (en) 2012-09-07 2017-08-08 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9724415B2 (en) 2012-09-07 2017-08-08 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9789185B2 (en) 2012-09-07 2017-10-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9782480B2 (en) 2012-09-07 2017-10-10 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9731008B2 (en) 2012-09-07 2017-08-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9731009B2 (en) 2012-09-07 2017-08-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9782479B2 (en) 2012-09-07 2017-10-10 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9737600B2 (en) 2012-09-07 2017-08-22 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9382317B2 (en) 2012-09-07 2016-07-05 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9757454B2 (en) 2012-09-07 2017-09-12 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9770507B2 (en) 2012-09-07 2017-09-26 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10058589B2 (en) * 2012-10-26 2018-08-28 Lupin Limited Stable pharmaceutical composition of Etanercept in a phosphate citrate buffer with glycine as an anti-aggregating agent
US20150283241A1 (en) * 2012-10-26 2015-10-08 Lupin Limited Stable pharmaceutical composition of tnfr:fc fusion protein
US9649383B2 (en) * 2012-11-19 2017-05-16 Merck Sharp & Dohme Corp. Liquid formulations for TNFR:Fc fusion proteins
US20150290325A1 (en) * 2012-11-19 2015-10-15 Merck Sharp & Dohme Corp. LIQUID FORMULATIONS FOR TNFR:Fc FUSION PROTEINS
WO2014099636A1 (en) * 2012-12-18 2014-06-26 Merck Sharp & Dohme Corp. Liquid formulations for an anti-tnf alpha antibody
US9844594B2 (en) 2012-12-18 2017-12-19 Merck Sharp & Dohme Corp. Liquid formulations for an anti-TNF α antibody
EP2934587A4 (en) * 2012-12-18 2016-08-03 Merck Sharp & Dohme LIQUID FORMULATIONS FOR ANTI-TNFALPHA ANTIBODY
WO2014114651A1 (en) * 2013-01-24 2014-07-31 Glaxosmithkline Intellectual Property Development Limited Tnf-alpha antigen-binding proteins
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
JP2014202667A (ja) * 2013-04-08 2014-10-27 株式会社島津製作所 抗体医薬用粒子径分布測定装置及び抗体医薬の粒子径分布測定方法
US20160129112A1 (en) * 2013-05-28 2016-05-12 Momenta Pharmaceuticals, Inc. Pharmaceutical Compositions Comprising Pyrophosphate
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
EP3057616A4 (en) * 2013-10-16 2017-06-14 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
AU2014337263B2 (en) * 2013-10-16 2019-12-12 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
US10376582B2 (en) 2013-10-16 2019-08-13 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
WO2015057910A1 (en) 2013-10-16 2015-04-23 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US20150299249A1 (en) * 2013-10-18 2015-10-22 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9315574B2 (en) 2013-10-18 2016-04-19 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9266949B2 (en) 2013-10-18 2016-02-23 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200069B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
CN104707146A (zh) * 2013-12-16 2015-06-17 浙江海正药业股份有限公司 一种含有阿达木单抗的药物组合物
US10918698B2 (en) * 2014-03-29 2021-02-16 Intas Pharmaceuticals Ltd. Lyophilized pharmaceutical composition of Fc-peptide fusion protein
US11752208B2 (en) 2014-05-23 2023-09-12 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11707524B2 (en) 2014-05-23 2023-07-25 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
EP2946765A1 (en) * 2014-05-23 2015-11-25 Ares Trading S.A. Liquid pharmaceutical composition
US10493152B2 (en) 2014-05-23 2019-12-03 Fresenius Kabi Deutschland Gmbh Adalimumab formulations
US11752209B2 (en) 2014-05-23 2023-09-12 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US11712471B2 (en) * 2014-05-23 2023-08-01 Fresenius Kabi Deustschland GmbH Liquid pharmaceutical composition
RU2719431C2 (ru) * 2014-05-23 2020-04-17 Фрезениус Каби Дойчланд Гмбх Жидкая фармацевтическая композиция
EP3939566A1 (en) * 2014-05-23 2022-01-19 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
AU2018220051B2 (en) * 2014-05-23 2020-06-04 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
EP3741358A1 (en) * 2014-05-23 2020-11-25 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
EP2946767A1 (en) * 2014-05-23 2015-11-25 Ares Trading S.A. Liquid pharmaceutical composition
US10426832B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
AU2020286276B2 (en) * 2014-05-23 2023-06-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
EP3476386A1 (en) * 2014-05-23 2019-05-01 Fresenius Kabi Deutschland GmbH Liquid pharmaceutical composition
EP3050557A1 (en) * 2014-05-23 2016-08-03 Ares Trading S.A. Liquid pharmaceutical composition
WO2015177057A1 (en) * 2014-05-23 2015-11-26 Ares Trading S.A. Liquid pharmaceutical composition
US10729769B2 (en) 2014-05-23 2020-08-04 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10772961B2 (en) 2014-05-23 2020-09-15 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10426833B2 (en) 2014-05-23 2019-10-01 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
WO2015177059A1 (en) * 2014-05-23 2015-11-26 Ares Trading S.A. Liquid pharmaceutical composition
WO2016057739A1 (en) * 2014-10-09 2016-04-14 Regeneron Pharmaceuticals, Inc. Process for reducing subvisible particles in a pharmaceutical formulation
US10342876B2 (en) 2014-10-09 2019-07-09 Regeneron Pharmaceuticals, Inc. Process for reducing subvisible particles in a pharmaceutical formulation
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
WO2016120413A1 (en) * 2015-01-28 2016-08-04 Mabxience S.A. Pharmaceutical formulations for anti-tnf-alpha antibodies
WO2016124588A1 (en) * 2015-02-06 2016-08-11 Ares Trading S.A. Liquid pharmaceutical composition
EP3053572A1 (en) * 2015-02-06 2016-08-10 Ares Trading S.A. Liquid pharmaceutical composition
EP3053573A1 (en) * 2015-02-06 2016-08-10 Ares Trading S.A. Liquid pharmaceutical composition
US20180296674A1 (en) * 2015-02-06 2018-10-18 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
US10179811B2 (en) 2015-04-10 2019-01-15 Fresenius Kabi Deutschland Gmbh Methods of treating Crohn's disease or ulcerative colitis using an induction dosing regimen comprising anti-TNF-alpha antibody
EP3078675A1 (en) 2015-04-10 2016-10-12 Ares Trading S.A. Induction dosing regimen for the treatment of tnf alpha mediated disorders
US10689440B2 (en) 2015-04-10 2020-06-23 Fresenius Kabi Deutschland Gmbh Method of treating Crohn's disease and ulcerative colitis by using an induction dosing regimen of adalimumab
US10669333B2 (en) 2015-04-10 2020-06-02 Fresenius Kabi Deutschland Gmbh Method of treating a tumor necrosis factor α (TNFα)-related disorder by using an induction dosing regimen of adalimumab
EP3078676A1 (en) 2015-04-10 2016-10-12 Ares Trading S.A. Induction dosing regimen for the treatment of tnf alpha mediated disorders
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
CN109563161A (zh) * 2016-02-03 2019-04-02 安口生物公司 用于提高抗体稳定性的缓冲制剂
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
WO2017136433A1 (en) * 2016-02-03 2017-08-10 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US11576971B2 (en) 2016-04-20 2023-02-14 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11071782B2 (en) 2016-04-20 2021-07-27 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11369896B2 (en) 2016-08-16 2022-06-28 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11571636B2 (en) 2016-08-16 2023-02-07 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11850535B2 (en) 2016-08-16 2023-12-26 Regeneron Pharmaceuticals, Inc. Methods for quantitating individual antibodies from a mixture
US11680930B2 (en) 2016-10-25 2023-06-20 Regeneron Pharmaceuticals, Inc. Methods and systems for chromatography data analysis
US11333642B2 (en) 2016-10-25 2022-05-17 Regeneran Pharmaceuticals, Inc. Methods and systems for chromatography data analysis
RU2665966C2 (ru) * 2016-12-30 2018-09-05 Закрытое Акционерное Общество "Биокад" Водная фармацевтическая композиция рекомбинантного моноклонального антитела к ФНОα
US10799597B2 (en) 2017-04-03 2020-10-13 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy
US11633476B2 (en) 2017-05-02 2023-04-25 Merck Sharp & Dohme Llc Stable formulations of programmed death receptor 1 (PD-1) antibodies and methods of use thereof
US11845798B2 (en) 2017-05-02 2023-12-19 Merck Sharp & Dohme Llc Formulations of anti-LAG3 antibodies and co-formulations of anti-LAG3 antibodies and anti-PD-1 antibodies
WO2019057631A1 (en) 2017-09-20 2019-03-28 Alvotech Hf PHARMACEUTICAL FORMULATIONS FOR ADALIMUMAB
US11884698B2 (en) 2018-07-02 2024-01-30 Regeneron Pharmaceuticals, Inc. Systems and methods for preparing a polypeptide from a mixture

Also Published As

Publication number Publication date
MX2011011772A (es) 2012-02-08
UY32609A (es) 2010-12-31
SG175188A1 (en) 2011-11-28
AU2010246168A1 (en) 2011-11-10
CA2760185A1 (en) 2010-11-11
IL215643A0 (en) 2012-01-31
NZ613809A (en) 2015-02-27
WO2010129469A1 (en) 2010-11-11
CN102458469A (zh) 2012-05-16
TWI480064B (zh) 2015-04-11
CN104490767A (zh) 2015-04-08
JP2012526121A (ja) 2012-10-25
TW201043263A (en) 2010-12-16
CN102458469B (zh) 2014-12-24
SG10201401995UA (en) 2014-08-28
AR076748A1 (es) 2011-07-06
WO2010129469A8 (en) 2012-02-23
KR20120038406A (ko) 2012-04-23
EP2427211A1 (en) 2012-03-14
RU2011149327A (ru) 2013-06-10
EP2427211A4 (en) 2013-05-01
TW201526923A (zh) 2015-07-16
NZ595694A (en) 2013-09-27
US20140141008A1 (en) 2014-05-22
US20140141007A1 (en) 2014-05-22
RU2560701C2 (ru) 2015-08-20

Similar Documents

Publication Publication Date Title
US20140141008A1 (en) Stable high protein concentration formulations of human anti-tnf-alpha-antibodies
US9302011B2 (en) Formulation of human antibodies for treating TNF-α associated disorders
JP6120452B2 (ja) 抗体製剤
AU2013204238A1 (en) Stable high protein concentration formulations of human anti-TNF-alpha-antibodies

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT BIOTECHNOLOGY LTD., BERMUDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT LABORATORIES;REEL/FRAME:024729/0934

Effective date: 20100714

Owner name: ABBOTT GMBH & CO., KG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FRAUNHOFER, WOLFGANG;REEL/FRAME:024729/0914

Effective date: 20100701

Owner name: ABBOTT GMBH & CO. KG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KRAUSE, HANS-JUERGEN;NEU, MICHAEL;REEL/FRAME:024729/0842

Effective date: 20100630

Owner name: ABBOTT BIOTECHNOLOGY LTD., BERMUDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT GMBH & CO. KG;REEL/FRAME:024729/0955

Effective date: 20100621

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FRAUNHOFER, WOLFGANG;REEL/FRAME:024729/0870

Effective date: 20100701

AS Assignment

Owner name: ABBVIE BIOTECHNOLOGY LTD., BERMUDA

Free format text: CHANGE OF NAME;ASSIGNOR:ABBOTT BIOTECHNOLOGY LTD.;REEL/FRAME:032131/0026

Effective date: 20120627

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION